Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Reviews

The principles of directed cell migration


Shuvasree SenGupta1, Carole A. Parent1,2,3,4 and James E. Bear 5,6 ✉

Abstract | Cells have the ability to respond to various types of environmental cues, and in many
cases these cues induce directed cell migration towards or away from these signals. How cells
sense these cues and how they transmit that information to the cytoskeletal machinery governing
cell translocation is one of the oldest and most challenging problems in biology. Chemotaxis,
or migration towards diffusible chemical cues, has been studied for more than a century, but
information is just now beginning to emerge about how cells respond to other cues, such as substrate-​
associated cues during haptotaxis (chemical cues on the surface), durotaxis (mechanical
substrate compliance) and topotaxis (geometric features of substrate). Here we propose four
common principles, or pillars, that underlie all forms of directed migration. First, a signal must be
generated, a process that in physiological environments is much more nuanced than early studies
suggested. Second, the signal must be sensed, sometimes by cell surface receptors, but also in
ways that are not entirely clear, such as in the case of mechanical cues. Third, the signal has to be
transmitted from the sensing modules to the machinery that executes the actual movement, a
step that often requires amplification. Fourth, the signal has to be converted into the application
of asymmetric force relative to the substrate, which involves mostly the cytoskeleton, but perhaps
other players as well. Use of these four pillars has allowed us to compare some of the similarities
between different types of directed migration, but also to highlight the remarkable diversity in
the mechanisms that cells use to respond to different cues provided by their environment.

Cell migration is critical in a wide array of physiological, microscopy8. In this scheme, the first event is the protru-
1
Department of Pharmacology, developmental and disease-​related processes, and basic sion of a leading edge, which in fibroblasts is dominated
University of Michigan Medical tenets governing this process have been uncovered over by lamellipodia and filopodia. Next, the cell generates
School, Ann Arbor, MI, USA. the years. In vivo, cells must be able to perceive a vari- initial adhesions with the substrate. These adhesions
2
Department of Cell and ety of cues in their environment and migrate towards or connect to the contractile machinery of actomyosin
Developmental Biology, away from these cues so as to execute morphogenetic stress fibres and, through a combination of pulling from
University of Michigan Medical
programmes during development, mount an immune the front and squeezing from the rear, the cell body
School, Ann Arbor, MI, USA.
response and repair damaged tissues. When this process moves forward. Finally, old adhesions are detached from
3
Rogel Cancer Center,
University of Michigan,
goes awry, devastating consequences often ensue. Failure the substrate or dissolved at the trailing edge. Although
Ann Arbor, MI, USA. of cells to migrate in the appropriate way can lead to textbooks describe this as a stepwise cycle of sequential
4
Life Sciences Institute, defects during neuronal development linked to cognitive steps, in reality, all events of the cycle occur simultane-
University of Michigan, deficits1, chronic wounds that never heal2 and immune ously and likely influence the other steps rather than
Ann Arbor, MI, USA. deficiencies3. Improperly initiated or misdirected cell proceeding as a series of independent events. In addi-
5
UNC Lineberger migration can be equally detrimental, leading to invasive tion, many of the concepts of the Abercrombie cycle
Comprehensive Cancer Center, metastatic cancer4, autoimmune disease5 and fibrosis6. are specific to mesenchymal cells such as fibroblasts on
University of North Carolina
at Chapel Hill School of
Biologists have studied the process of directed migra- 2D surfaces and may not apply to other cell types that
Medicine, Chapel Hill, tion for more than a century, but many mysteries remain use different modes of migration or during migration
NC, USA. about how this process works at a mechanistic level. in different physiological environments (see Box 1 for a
6
Department of Cell Biology To define how cells move directionally towards var- primer on different modes of migration).
and Physiology, University ious cues, it is critical to understand the basics of cell In this Review, we present a four-​part conceptual
of North Carolina at Chapel migration. Perhaps the most influential paradigm for framework for understanding directed cell migration
Hill School of Medicine,
Chapel Hill, NC, USA.
describing cell migration is the four-​step cycle of cell towards a variety of cues, including diffusible chemical
✉e-​mail: crawling developed by Michael Abercrombie, an early cues (chemotaxis), chemical cues on a surface (hapto­
jbear@email.unc.edu pioneer of the field7. This paradigm arose from his taxis), mechanical substrate compliance (durotaxis),
https://doi.org/10.1038/ observations of migrating primary and cultured fibro- geometric features of the substrate (topotaxis; also known
s41580-021-00366-6 blasts by phase contrast and interference reflection as contact guidance) and electric fields (galvanotaxis;

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 529

0123456789();:
Reviews

Box 1 | Modes of cell migration and plasticity all aspects of this large topic. Thus, we will avoid descrip-
tions of new technologies for generating signal gradients
Cells can migrate singly or collectively as groups227–229. Historically, single-​cell migration or for quantifying migration or mathematical/theoretical
has been divided into mesenchymal and amoeboid modes of migration, although models of this process, although we point to a few appro-
these classifications are complicated by the plasticity of migration modes in different
priate reviews of these topics (see refs9,10). Instead, we
environments (see below). Fibroblasts, various stem cells and some cancer cells often
will focus on how new findings provide insight into the
use the mesenchymal migration mode, which is defined by several characteristics, such
as a strong dependence on adhesion to the extracellular matrix (ECM), an elongated underlying principles of directed migration and suggest
morphology in 3D environments, actin-​based protrusions such as lamellipodia or questions to be addressed by future studies.
filopodia at their leading edge, and the ability to generate strong traction forces on the
substrate through contractile actin networks. These characteristics usually give rise to Four pillars of directed migration
slower migration velocity93. The amoeboid migration mode is used by a wide range of To organize the large amount of information necessary
cells, including primordial germ cells, single-​cell social amoebas such as Dictyostelium to understand directed migration in response to vari-
discoideum and immune cells such as leukocytes230. In this mode, cells exhibit a more ous cues, we developed a generic, conceptual frame-
rounded morphology, undergo constant shape changes through rapid extension and work of four events that must occur during all forms
retraction of membrane protrusions, and engage with the substrate through weak of directed mig­ration, which we term the ‘four pillars of
adhesions, which usually lead to higher migration velocity. Amoeboid protrusions
directed mig­ration’ (Table 1). The pillars include genera­
vary from lamellipodia and filopodia driven by actin polymerization to actin-​free
transient spherical blebs that rely on myosin-​based contraction and pressure-​driven ting the signal, sensing the signal, transmitting the signal
cytosolic flow217,230. and executing the signal. For each pillar, we separate the
Cells can also move collectively as groups, which presents its own challenges and various forms of directed migration by cue except for
opportunities. Both epithelial and mesenchymal cells exhibit collective migration, the fourth pillar, where we consider how the various sig-
which is important for tissue remodelling during morphogenesis, wound closure and nalling mechanisms converge on a common set of cell
cancer cell invasion211,229. During collective migration, signals from external cues are translocation machineries.
transmitted to the entire mass of cells through the integration of intracellular and
intercellular signalling cascades as well as mechanotransduction at cell–cell junctions Generating the signal
and cell–ECM interfaces212. This results in front–rear polarity at a supracellular level. For directed migration to occur, a signal must first be
A group of cells situated at the front of the supracellular unit generally becomes the
generated. This signal may be a transient cue such as
leader cells in response to external cues and extend stable lamellipodia or filopodia
towards the substrate, whereas the follower cells situated at the rear extend a diffusible chemical signal secreted into the environ-
small transient cryptic lamellipodia. The stable protrusions possibly together with ment meant to direct cells for a short burst of migra-
the transient ones promote the formation of focal adhesions with the ECM to exert tion. Alternatively, the signal may involve a long-​lasting
traction forces towards the substrate. Application of these forces physically deforms change to the environment that guides cells for an
the matrix, creating a path for the entire cohort211,229. Leader cells can also secrete extended time, such as the generation of physical paths.
matrix metalloproteinases that remodel the surrounding ECM, paving the way for
collective migration during cancer invasion. In addition to this traction-​based collective Chemotaxis. Chemotaxis is mediated by the generation
migration, cells can adopt a propulsion-​based motility analogous to the amoeboid of diffusible cues. When these cues are presented uni-
mode detected, for instance, in colon cancer cell clusters231. formly, cells undergo chemokinesis, where they migrate
Complicating any classification scheme of cell migration is the fact that cells can
randomly with either higher speed and/or higher turn-
switch between modes of single-​cell migration and between single-​cell and collective
migration, depending on a variety of factors, such as tissue topology, ECM composition ing frequency relative to unstimulated cells11. However, if
and the degree of adhesion to it, as well as the presence of biochemical cues232,233. the promigratory signal is presented in the form of a gra-
For example, physical confinement and low adhesion enable slow-​moving fibroblasts dient, directed migration occurs12 (Fig. 1a). The diffusible
and epithelial cells to transition into a faster migration mode, where large stable blebs agents that induce directed migration include a large and
are favoured by high cell contractility232. Similar stable bleb-​based fast migration is diverse group of chemoattractants produced by different
observed in zebrafish progenitor cells induced by spatial confinement in vitro233 and sources. These comprise formylated peptides13, products
in vivo at transplantation-​induced wound sites of the embryo, where the cortical of the complement cascade14, phospholipid metabolites15
contractility is elevated. Confinement-​associated plasticity in the migration mode is and a large family of chemokines and growth factors
also detected in leukocytes. For example, neutrophils with a genetically disrupted that are derived from endothelial, epithelial and stromal
branched actin network switch from multiple finger-​like protrusions to smooth
cells16. In addition, ATP and hydrogen peroxide have
bleb-​based leading-​edge protrusions when exposed to confined microenvironments234.
Cancer cells can also adopt a rounded, bleb-​based migration mode in low-​adhesion 3D been reported to act as autocrine signals to amplify
environments or when their matrix metalloproteinase activities are disrupted. In addition, chemotactic signals 17–19. Furthermore, specialized
physicochemical parameters such as hypoxia, which is a prominent feature of solid secreted proteins, such as Slits, netrins, semaphorins
tumours, have been shown to promote the transition of collectively invading cancer and ephrins, are well-​known axon guidance cues20–23.
cells into individually moving amoeboid cells, and enhanced cancer dissemination235. The diverse biochemical nature of these chemotactic
The extreme plasticity in migration modes displayed by cancer cells may allow them cues, with distinct diffusion coefficients and affinities for
to adapt to many tissue environments and contribute to disease progression. their cognate receptors, presents considerable challenges
for the generation and maintenance of stable gradients
also known as electrotaxis) (Table 1). This framework is during chemotaxis.
Lamellipodia
Broad, sheet-​like protrusions deliberately generic to facilitate comparisons and con- A gradient can be established by simple diffusion from
that contain branched trasts between different types of migration-​inducing cues a source or by regulating the removal of the attractant24.
and linear actin filaments. and migration modalities. By comparing different forms Examples of mechanisms cells use to regulate gradient
A variety of cell types, including of directed migration, we highlight the progress made in formation and avoid receptor saturation (a situation
fibroblasts, neural crest cells and
macrophages, use lamellipodia
the field and reveal gaps in our understanding of mole­ when cells are no longer able to perceive concentration
to explore longer distances cular underpinnings driving the directionality of cell differences of the chemoattractant) include degrada-
through the extracellular matrix. migration. This Review cannot comprehensively cover tion of chemoattractants by enzymatic or proteolytic

530 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

Filopodia
breakdown, and endocytosis of cell surface-​b ound self-​generating gradients have recently been proposed
Finger-​like protrusions that chemoattractants via scavenger/decoy receptors that as an alternative mechanism to generate chemical gradi-
contain bundles of linear specifically remove their ligand without initiating cell ents. In this case, migrating cells would secrete enzymes
F-​actin. Filopodia serve to polarity/migration signalling (reviewed in25) (Fig. 1b). For that break down chemoattractants initially distributed
probe local environmental
cues, provide directionality
example, it was established that a negative-​feedback loop uniformly — as observed for Dictyostelium discoideum
and maintain persistence between CXCL12 and its receptor CXCR7 is required to cells migrating towards folic acid28, and melanoma and
of migrating cells. maintain optimal CXCL12 concentration in the zebrafish pancreatic cells responding to lysophosphatidic acid29,30.
posterior lateral line primordium26. Using a clever synthetic Such a mechanism can theoretically give rise to steep
Stress fibres approach, where GFP is used to generate diffusible gra- gradients that work over long distances and convoluted
Contractile arrays of actin
and non-​muscle myosin II
dients, it was recently shown that combining the expres- migratory pathways. Accordingly, with use of artificial
that are mechanically coupled sion of non-​signalling decoy receptors with receptors complex environments and mathematical modelling, it
to the substrate through engineered to respond to GFP allows the synthetic GFP was recently shown that the breakdown of attractants
integrin-​based focal adhesions. gradient to generate normal growth and patterning of allows D. discoideum and pancreatic metastatic cell
the Drosophila melanogaster wing pouch27. In addition, lines to navigate long, complex paths in a manner that

Table 1 | The four pillars of directed cell migration


Migration Cue Signal generation Signal sensing Signal transmission Signal execution
mode
Chemotaxis Diffusible chemical Simple diffusion GPCRs Classical signalling Leading-​edge
released from pathways involving small protrusions (all types)
Regulated removal RTKs
cells or deposited and large G proteins
by degradation of the Localized regulation
extracellular vesicles Other receptors, such as
chemoattractant or decoy PI3K of non-​muscle myosin
axon guidance receptors
receptors II and contractility
TORC2
Release of extracellular
PLA2
vesicles
MAPK/ERK
Haptotaxis Substrate-​bound ECM secretion and For ECM, integrins, Classical integrin signalling Biased protrusion
chemical cues such deposition but different adhesion pathways: Rho-​family generation through
as an immobilized structure impacts GTPases, FAK–Src, etc. a positive-​feedback
Binding of soluble factors
chemokine or ECM signalling outcome loop. Requires the
to a substrate (mostly Bound chemokine:
ECM) For substrate-​bound probably same as diffusible Arp2/3 complex
Exposing new sites on chemokines, regular cue
the substrate through receptors, but signalling
enzymatic action kinetics may be different
for different receptor–
ligand pairs
Durotaxis Differential substrate Passive: creating a stiff Integrins Unclear, but two Similar to other
compliance substrate by crosslinking mechanisms have forms of migration
Membrane tension
of ECM components or been proposed: role of but biased relative
and/or invagination
ECM deposition pure mechanics using to stiffness gradient
Focal adhesion actomyosin system
Active: cells or tissues
components or the involvement of
exerting a force on the
substrate that is sensed Actomyosin filaments mechanically triggered
by other cells signalling events
LINC complex
Topotaxis Geometric Preformed tunnels Cells adhere and conform Cell and nuclear shape Topology/
properties of the created by other cells to the topology and/or change may affect geometry biases
migration substrate the geometry of the both signalling and the force-​generating
Trails created by
irrespective of migration substrate with cytoskeleton but the mechanisms of actin
proteolytic ECM
mechanical or the help of focal adhesion mechanisms remain polymerization
remodelling
chemical properties components unclear and actomyosin
Topological features contractility
created by non-​lytic ECM Membrane curvature-​
deformation sensing proteins

1D fibrils such as bundles Nucleus deformation


of collagen
Topology of natural tissue
elements
Galvanotaxis Electric fields Ionic differences Electrophoretic Clustering of membrane Similar to chemotaxis
generated by movement of charged proteins/lipids must but biased relative to
transepithelial barriers surface proteins and activate signalling, but charge
such as in the skin, lipids within the plane the mechanisms remain
disrupted by wounding of the membrane unclear
Arp2/3 complex, actin-​related protein 2/3 complex; ECM, extracellular matrix; FAK, focal adhesion kinase; GPCR, G protein-​coupled receptor; LINC complex,
linker of nucleoskeleton and cytoskeleton complex; PI3K, phosphoinositide 3-​kinase; PLA2, phospholipase A2; RTK, receptor tyrosine kinase; TORC2, target of
rapamycin complex 2.

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 531

0123456789();:
Reviews

a Signal generation
Chemotaxis Haptotaxis Durotaxis

Chemoattractant Collagen Low stiffness High stiffness


fibre
LOX

ECM protein
Chemoattractant
Enzyme Crosslinked ECM fibres
Chemical Release of ECM-bound
concentration chemoattractant
Topotaxis Galvanotaxis

–––––
Tissue Electric current
Confined tunnel Nanostructures wounding

MMP +++ +++ –––––


Tracks of Tracks generated by Transepithelial Electric field
aligned fibres proteolytic remodelling electric potential generation

b Gradient generation and stabilization


Through enzymes and decoy receptors Through EV release

Enzyme
GPCR

Endosome
Multivesicular
body with EVs
Decoy
receptor
EV establishing EV establishing
heterotypic homotypic gradient
gradient

Fig. 1 | Generating the signal. a | The diverse ways by which cues for directional migration are generated. During chemotaxis,
soluble chemoattractants released from bacteria or cellular sources diffuse to form chemical gradients. During haptotaxis,
extracellular matrix (ECM) proteins and chemokines released from cellular sources are deposited onto the ECM and
generate gradients of immobilized chemical cues. In some cases, ECM-​bound chemokines are released from the matrix
Substrate compliance
by cellular proteolytic activities (scissors) and provide soluble cues for chemotaxis. During durotaxis, gradients of stiffness
The mechanical resistance
can be generated by lysyl oxidase (LOX)-​mediated ECM crosslinking. During topotaxis, the geometry of the existing tissue
provided by non-​rigid
substrates (for example,
structures, aligned fibres or tracks generated by proteolytic remodelling (via matrix metalloproteinases (MMPs), scissors)
collagen gels) to the contractile or deformation provides directional signals. During galvanotaxis, electric fields generated at wounding sites as a result
forces exerted by cells as they of the loss of transepithelial potential provide guidance cues for cells involved in damage repair. b | Cartoon explaining
engage the substrate. how stable gradients are generated and maintained during chemotaxis. Uniformly present soluble chemicals are either
degraded by enzymes or scavenged (via endocytic internalization) by decoy receptors to establish a gradient (left).
Complement Cells release extracellular vesicles (EVs), such as exosomes carrying either identical (homotypic gradient) or distinct
Complement proteins are (heterotypic gradient) chemical cues, to generate a stable secondary gradient (right). GPCR, G protein-​coupled receptor.
products of the complement
pathway generally activated
as part of the innate immune is dependent on attractant diffusibility, cell speed and that harbour motifs that bind to extracellular matrix
response to infection. Some path complexity31. (ECM) components and can be later released by
complement proteins, such as
C5a, act as chemoattractants
Gradients can be propagated by means other than cell-​mediated proteolysis of the ECM component33.
that guide leukocytes to sites simple diffusion 32. For instance, morphogens have Similarly, it has been shown that neutrophils migrat-
of infection. long been known to be secreted in precursor forms ing in 3D collagen matrices activate discoidin domain

532 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

receptor 2 (DDR2), which binds collagen I, and induces of cells can secrete ECM proteins, such as fibronectin,
the secretion of matrix metalloproteinases and the laminin and various collagens, into the environment to
Posterior lateral line release of collagen-​derived chemotactic peptides that form insoluble arrays that become migration substrates
primordium act in an autocrine manner to stabilize neutrophil or bind to existing substrates, thereby functioning as
A group of cells that migrate directionality34. The concept, referred to as ‘autologous migration cues for the cells themselves or for other cells
together from the ear to the tip
of the tail of zebrafish as they
chemotaxis’, has also been reported to be involved dur- in the vicinity49,50. Unlike diffusible chemotactic cues,
periodically deposit primary ing the CCR7-​driven directed migration of tumour cells. ECM haptotactic cues tend to be relatively stable and
neuromasts. In this case, chemoattractant gradients were generated long-​lasting. As many components of the ECM can bind
from autocrine signals as a result of interstitial flow that to each other, their deposition can be iterative to create
Morphogens
produced advection fields35,36. complex mixtures of haptotactic cues51. In addition, cells
Signal molecules that originate
from a tissue and diffuse to During development, the transportation of signal- can locally degrade ECM components to further sculpt
generate a concentration ling molecules along filopodium-​like protrusions called remarkably complex migration environments52.
gradient. Morphogens exert ‘cytonemes‘ or ‘tunnelling nanotubes’ (thin cellular The specific molecular mechanisms of ECM secre-
long-​range signalling effects protrusions involved in cell–cell communication) or tion remain poorly understood. Recent work suggests
important for growth and tissue
patterning during development.
through transcellular transport (transcytosis) has been that caveolin-​dependent regulation of exosome biogen-
reported to be involved in the generation of mitogen esis is a key step in ECM secretion and deposition in
Advection fields gradients37,38. There is also evidence that the packaging fibroblasts53, but the universality of this mechanism will
Fluid flows such as interstitial of chemoattractants in extracellular vesicles (in particu- need to be explored in other cell types. Furthermore, ini-
flow in tissues that can create
lar exosomes) importantly contributes to the genera- tial cell engagement with the ECM can trigger the depo-
an advection field or directional
transfer of molecules in the tion of gradients during chemotaxis. In D. discoideum sition of fibronectin at the leading edge of cells through
liquid phase around cells, cells, extracellular vesicles have been shown to contain a mechanism involving one of the Rho-​family GTPases,
which in turn can create the machinery to synthesize and release the chemo­ CDC42 (ref.54). This creates a positive-​feedback loop for
asymmetries in secreted attractant cAMP; these extracellular vesicles mediate the cells to essentially lay down tracks and facilitate persis-
autocrine chemoattractants,
leading to autologous
relay of chemotactic signals during chemotaxis and tent migration on ECM compositional gradients. Cell
chemotaxis. Advection fields the alignment of cells in a head-​to-​tail fashion in a process engagement with the ECM can also modify the structure
can also form in the cytoplasm. referred to as ‘streaming’39,40. In macrophages and den- of the array, whereby integrin-​dependent cell contacts
dritic cells, the secondary chemoattractant leukotriene induce the reorganization of fibronectin fibrils55. An
Extracellular vesicles
B4 (LTB4) has been reported to be present in exosomes interesting pathophysiological example of ECM depo-
A group of heterogeneous
vesicles (several nanometres to and promote migration41. In neutrophils, LTB4 is released sition occurs in the retina of patients with diabetes or
micrometres in size) that carry in response to primary chemoattractant stimulation and macular degeneration, where inappropriate deposition
a variety of cargos, including acts in an autocrine and paracrine fashion to stabilize of fibronectin leads to thickening of Bruch’s membrane
proteins, lipids and nucleic neutrophil cell polarization and to relay signals to dis- (the innermost layer of the retina) and inappropri-
acids, and are secreted by
cells to the extracellular
tant neutrophils42,43. Similarly, dendritic cell migration ate neovascularization56. Such increased deposition of
space to facilitate cell–cell was recently reported to depend on exosomes released ECM proteins could generate abnormal haptotactic cues.
communication. from lymphatic endothelial cells in a CX3CL1 (also However, increased ECM deposition will also change
known as fractalkine)-​dependent fashion44. Moreover, the mechanical landscape of the microenvironment,
Exosomes
chemokine-​containing exosomes isolated from stressed which could trigger a pathological durotactic response
The smallest subtype of
extracellular vesicles, with tumour cells have been reported to activate and induce as described later.
a size ranging from 50 to the migration of T cells45, and neutrophils have been In addition to ECM haptotactic cues, cells secrete
150 nm. Exosomes are shown to package and release CXCL12 via secretory factors such as chemokines or other guidance factors
generated as intraluminal vesicles, leaving behind CXCL12-​containing trails that that bind tightly to existing ECM arrays57, thus gen-
vesicles which are secreted to
the extracellular space when
attract T cells to infection sites46. In these contexts, the erating haptotactic cues that are sensed by direct cell
intraluminal vesicle-​carrying packaging of chemotactic cues in extracellular vesicles/ engagement rather than acting at a distance through
multivesicular bodies fuse with exosomes is poised to protect attractants from harsh diffusion like during chemotaxis. Generation of these
the plasma membrane. extracellular environments, degradation and/or rapid cues is regulated by mechanisms similar to the ones
diffusion. Furthermore, we envision that as the vesicles responsible for the generation of diffusible, chemotactic
Caveolin
Integral membrane protein are deposited, they can persist after the cells have left cues. Nevertheless, as noted already, such ECM-​bound
family required for flask-​ the area and continue to deliver chemotactic cues to chemokines can also be released to switch a haptotactic
shaped (caveola) membrane generate long-​lasting secondary gradients to recruit dis- cue into a locally functioning chemotactic cue58,59.
structure formation. Caveolins tant cells to sites required for their action (such as sites
are also involved in membrane
trafficking, exocytosis,
of inflammation for leukocytes) (Fig. 1b). In addition, Durotaxis. In addition to sensing molecular cues, cells
endocytosis, extracellular extracellular vesicles have also been reported to mediate have the ability to sense differences in substrate stiffness
vesicle formation and directional migration by regulating cell–ECM adhesion and respond by migrating towards or away from areas
extracellular vesicle assembly in tumour cells. In this case, autocrine secre- of higher stiffness (Fig. 1a). Such stiffness gradients have
cargo selection.
tion of fibronectin-​coated exosomes at the leading edge recently been demonstrated in vivo in the embryonic
Rho-​family GTPases of cells expressing fibronectin receptors allowed them to mouse limb bud60. The generation of these durotactic
A family of small proteins that establish connections to the ECM, which became coated cues requires changes in the mechanical environments
bind GDP or GTP and regulate with these extracellular vesicles47,48. that cells encounter, which can persist for long periods
a wide array of downstream and influence the migration of cells for days or much
signalling events. CDC42, Rac,
and RhoA are widely studied
Haptotaxis. Haptotaxis is the sensing of surface-​bound longer. For example, increased amounts of ECM deposi-
members of this family chemical cues (Fig. 1a). A primary haptotactic cue is tion can often lead to changes in the mechanical proper-
of proteins. provided by the components of the ECM. A variety ties of the local microenvironment, which can produce a

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 533

0123456789();:
Reviews

hybrid haptotactic–durotactic–topotactic cue61. Another Galvanotaxis. The existence of electric fields around
means by which mechanical cues can be generated is cutaneous wound sites has been known since the
through mechanical modification of the existing ECM, 1840s76, and these fields can serve as cell migration cues
either stiffening it or relaxing it. For example, the lysyl in a process called ‘electrotaxis’ or ‘galvanotaxis’ (Fig. 1a).
oxidase enzymes (LOXL1–LOLX4) can crosslink col- During the wounding process, the electric poten-
lagen fibrils and other ECM components to render a tial maintained by transepithelial resistance is short-​
stiffer network62,63. Interestingly, this group of enzymes circuited, and the resulting electric field can reach up
is frequently misregulated in cancer and other disease to 10 μA cm−2, a value in the range that can be sensed by
states such as during fibrosis, speaking to the importance cells77. In addition to wounds, electric fields have been
of mechanical control of the cell’s environment62. These documented during embryogenesis through transep-
mechanical changes can influence the proliferation and ithelial ion transport78. These transient electric fields
migration of tumour cells64 as well as surrounding stro- are sensed by cells in their vicinity, often provoking a
mal cells such as endothelial cells in the vasculature65. directed migration response.
Conversely, matrix-​degrading enzymes such as matrix
metalloproteinases can relax or soften the environment Sensing the signal
to further sculpt the mechanical landscape encountered Once a signal has been generated, cells must be able to
by cells66,67. sense the signal. In the case of chemotaxis, this is a fairly
straightforward process of receptor–ligand interactions.
Topotaxis. Topotaxis is driven by biophysical cues, However, the sensing step for other cues, such a mecha­
where cells sense the topographical features of the sur- nical compliance of the substrate, is less straightforward
rounding microenvironment. Natural tissue elements, and involves more complex mechanotransduction path-
including aligned collagen fibres, muscle strands, nerve ways engaging both surface proteins and mechanically
fibres, vascular tracks and pores or tunnel-​like confined coupled intracellular proteins.
trails within the ECM, often provide an anisotropic sur-
face architecture at nanometre or micrometre scale68,69 Chemotaxis. The mechanisms underlying how cells
(Fig. 1a). Migrating cells tend to adapt their shape to sense extracellular cues are, by far, best understood for
the available geometry of the surrounding substrate chemical cues in the context of chemotaxis (Fig. 2a).
to migrate in a preferred direction. However, it is impor- Work using D. discoideum in the late 1980s first identi-
tant to consider the cell types (taking into account their fied G protein-​coupled receptors (GPCRs) as the surface
unique properties) as well as the size of the confining receptor responsible for sensing cAMP — the main chemo­
space when one is reflecting on how topological cues pre- attractant for D. discoideum chemotaxis79. This work
scribe directional choices to migrating cells. For instance, was followed by a flurry of reports in the early 1990s
it has been shown that migrating leukocytes, which showing that chemokines are also sensed by GPCRs80–83.
exhibit amoeboid movement, follow pre-​existing trails More than 50 distinct chemokines have been identified
in 3D reconstituted collagen matrices70,71. Unlike tumour in humans and are grouped into the CL, CCL, CXCL
cells and fibroblasts, these leukocytes do not actively or CX3CL subfamily, depending on the sequential posi-
break down the ECM. Instead, leukocytes migrating in tioning of highly conserved cysteine residues84,85. These
interstitial tissue undergo a robust shape change guided are recognized by ~20 known conventional chemokine
by the matrix that induces transient deformation of the receptors, referred to as ‘CCRs’ or ‘CXCRs’, that share
collagen network, and squeeze through the preformed 25–80% sequence identity and exhibit the ability to
trails of larger pore size and least resistance. By contrast, bind multiple chemokines within a given chemokine
cancer cells during tissue invasion frequently depend on subfamily83,86,87. In addition, some chemokines can bind
proteolytic remodelling of the ECM to create their own to atypical chemokine receptors, which are structurally
trails, especially when encountering environments with related to conventional chemokine receptors but do
limited space and more resistance 69,72. However, non-​ not couple to signalling modules. Finally, formylated
proteolytic strategies, including ECM deformation, have peptides88, products of the complement cascade16, phos-
been reported in cancer cells with amoeboid features as pholipid metabolites89 and the small molecules ATP and
they make their way through tissue73. Depending on the ADP17,90 are all known to bind to GPCRs to mediate
tumour type, a customized migratory approach may their chemotactic activities, making GPCRs the main
also exist, where tumour invasion relies on protease-​ molecular chemotactic sensors.
G protein-​coupled receptors
(GPCRs). A family of plasma
dependent tunnel formation in the matrix, which guides Receptor tyrosine kinases (RTKs) that interact with
membrane receptors composed the migration of leading tumour or non-​tumour stromal growth factors, such as EGF and PDGF, are a second
of seven transmembrane cells (including fibroblasts, endothelial cells and smooth class of receptors that mediate chemotaxis91. Whereas
domains that couple to muscle cells), whereas the follower cells are simply car- GPCR-​mediated chemotactic signalling prevails in the
heterotrimeric G proteins to
ried along those tunnels without the need for active ECM context of amoeboid chemotaxis, mesenchymal cells
regulate responses mediated
by a variety of external signals. remodelling74. Further, it has been reported that cancer often use RTK-​mediated signalling during directional
cells during invasion can orient themselves parallel to migration, perhaps reflecting fundamental differences
Axon growth cone different topological features of the surrounding tissue, in physiological and environmental conditions encoun-
Motile structure at the tip and migrate directionally without requiring major ECM tered by each cell type92,93. In addition, axon growth cone
of growing axons that guides
directed extension of the axon
remodelling, suggesting that cells are capable of sensing guidance, which is required for patterning the nervous
and is important for patterning complex topological features of the microenvironment system, is mediated by several families of transmem-
of the nervous system. and tune their migratory response accordingly75. brane receptors that bind to secreted guidance cues

534 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

a Signal sensing
Chemotaxis Haptotaxis Durotaxis

Chemoattractant
receptor Chemoattractant
Chemoattractant receptor Chemoattractant LINC
receptor complex
Integrins
Immune Fibroblast Mechanosensing
cell components

Microtubules ECM protein


Collagen Crosslinked ECM fibres
Neuronal fibre
growth cone
Chemoattractant
receptor
Topotaxis Galvanotaxis
Protrusion–ECM
fibre alignment Electric current
Curvature
sensing BAR-family
Focal protein
adhesion
Tracks of aligned fibres Nanostructures +++ –––––
Shape Nuclear
adaptation deformation
Receptor clustering
via electromigration
Signal
Confined tunnel transduction

b Detailed machinery of durotaxis signal sensing

LINC
Membrane invagination complex Actin filament
during endocytosis Myosin
Mechanosensing
components Calcium ion
Stretch-activated
ion channel (Piezo)
Focal adhesion complex

Crosslinked ECM fibres

Fig. 2 | Sensing the signal. a | Various ways cells sense directional cues. During chemotaxis, cells sense the signal through
surface receptors (G protein-​coupled receptor (GPCRs), receptor tyrosine kinases or other transmembrane receptors),
which bind the soluble chemical cues. During haptotaxis, cells detect surface-​bound cues through integrin receptors and
GPCRs. During durotaxis, substrate stiffness is sensed by an array of mechanically coupled components located on the cell
surface, in the cytosol or at the nuclear envelope. During topotaxis, cells detect the geometry of available space and adapt
their shape by changing the orientation of membrane protrusions in parallel to the aligned extracellular matrix (ECM)
fibres, sensing topology-​induced membrane curvature by BAR-​family proteins, or gauging nuclear deformation resulting
from compression and shape change. During galvanotaxis, the electric field is sensed by electromigration of membrane
components (including signalling receptors) towards the cathode (+++) or the anode (− − − − −). b | Molecular machinery
for durotactic sensing. Cells sense gradients of stiffness using mechanosensors at the cell surface (including integrin
receptors in focal adhesions, invaginated membranes and stress-​activated ion channels), inside the cytoplasm (including
components of focal adhesions, actin filaments, microtubules and other mechanosensitive proteins) or at the nucleus
(LINC complex).

(such as Robo–Slit proteins or netrin–netrin receptors) Haptotaxis. Haptotactic cues are also sensed by cognate
and mediate either attractive or repulsive responses94–96. cell surface receptors (Fig. 2a). Integrins are used to sense
Neuronal guidance cues are also known to regulate haptotactic gradients composed of ECM components,
immune responses97, where, again, they can inhibit or where the spatial organization or clustering of integrins
promote migration98–100. is key to their signalling properties101. Numerous studies

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 535

0123456789();:
Reviews

have shown that small, nascent adhesions at the leading adhesions have been shown to be mechanically sensi-
edge of migrating cells recruit a different subset of sig- tive, including talin116, vinculin117 and p130Cas118. When
nalling and mechanical effectors compared with larger, focal adhesions are under differential mechanical load,
maturer adhesions further back under the cell body102. many of these proteins show altered conformation
Moreover, these small, nascent adhesions are critical for and/or altered interactions with other cytoplasmic com-
ECM haptotaxis through a specific signalling pathway ponents. However, many cells lack the large, stable focal
described in the next section. In the case of surface-​ adhesion structures of mesenchymal cells, where these
bound chemokines such as CCL21, the same GPCR phenomena have been mostly studied, and still display
(CCR7) that senses the diffusible version of this cue is sensitivity to substrate rigidity119. This suggests that
used to sense immobilized gradients103. Interestingly, focal adhesions may not be a universal durotactic sens-
dendritic cells following surface-​immobilized CCL21 ing structure across all cell types. Regardless of whether
gradients require non-​linear, exponential gradients for integrins are clustered in classic focal adhesion struc-
haptotaxis and do not undergo haptotaxis on linear sur- tures, they are still mechanically coupled to actin fila-
face gradients, suggesting differences in how the same ments, and several recent studies have shown that actin
cue is sensed when it is diffusible or surface-​bound. filaments themselves are sensitive to mechanical load by
In theory, cells could use other receptors such as cad- differentially binding proteins containing LIM domains
herins at cell–cell junctions to perceive haptotactic cues depending on the mechanical conditions (varying
presented by other cells, but this has yet to be reported. load)120,121. This differential response to mechanical load
Cell–cell junctions
could serve as a type of ‘durotactic receptor’, leading to
Stable or dynamic sites where
borders of two neighbouring Durotaxis. The mechanism of durotactic sensing is a altered cytoskeletal dynamics and structure, as well as
cells contact each other. matter of intense recent interest (reviewed in104–106). alteration of signalling pathways. Recent work has also
Cell–cell adhesion receptors Unlike the cell-​impermeant chemical cues driving shown that a subset of microtubules originating from the
and recruited adaptor proteins chemotaxis and haptotaxis, which must be sensed by Golgi apparatus are critical for durotaxis by regulating
are mechanically coupled to
the actin cytoskeleton.
cell surface receptors, mechanical force is not limited focal adhesion dynamics122.
by membranes. Thus, the ‘receptors’ or ‘sensors’ for duro­ Finally, actin filaments and microtubules are con-
Focal adhesions taxis could theoretically be on either side of the plasma nected to the nucleus by the mechanically sensitive
Multiprotein assemblies that membrane or even much deeper in the cytoplasm of the LINC complex123,124, which could function as a sensor for
physically connect extracellular
cell, as long as these components are mechanically cou- differential mechanical load on either side of the nucleus.
matrix components to the
intracellular actin cytoskeleton pled to the substrate. We envision that durotaxis results Indeed, differential positioning of the nucleus relative to
through integrin clusters. from the ensemble activation of multiple mechanically the rest of the cell has been linked to regulation of cell
Integrin-​mediated adhesion sensitive ‘receptors’ that act in concert to drive directed polarization during scratch-​induced migration of epi-
to extracellular matrix ligands cell migration. thelial monolayers125 as well as cell locomotion through
recruits a plethora of signalling
(Src and FAK) and structural
Several candidate durotactic ‘receptors’ have been piston-​like generation of pressure gradients in fibroblasts
(talin, paxillin and vinculin) identified (Fig. 2b) . Working from outside the cell migrating in a lamellipodium-​independent manner in
molecules to focal adhesions. inward, the first candidate receptor are integrins107,108. certain 3D environments126 (see the section Executing
Large, mechanically engaged Several recent biophysical studies have demonstrated the signal for details).
focal adhesions play a crucial
that these surface proteins are sensitive to mecha­
role in sensing mechanical
cues, while smaller, nascent nical load and are concentrated in structures relevant Topotaxis. We are only beginning to understand the
adhesions are critical for for cell migration, such as filopodia, lamellipodia and mechanisms for sensing topology (Fig. 2a). Similarly to
sensing haptotactic cues. focal adhesions109,110. However, as some cells can migrate durotaxis, sensors of topological cues are mechanically
by integrin-​independent mechanisms111, if they are coupled elements that are located on either side of the
Traction force
The stress vector at the
sensing substrate compliance, they may be using an cell surface or within internal compartments, such as
interface between a migrating integrin-​independent mechanism. Another potential the nucleus. Some of the sensing elements for topotaxis
cell and its substrate. source of mechanical sensing is the membrane itself, (for instance, focal adhesions) may overlap with those
which seems to prominently involve invaginations of for both haptotaxis and durotaxis. Over the past dec-
LIM domains
the membrane formed during either clathrin-​based ade, a large number of studies have monitored changes
Protein structural domains
named after the proteins or caveolin-​based endocytosis. Indeed, several studies in cell shape, protrusion shape, actin cytoskeleton and
LIN-11, ISL1 and MEC-3. have shown that these endocytosis pathways display motility in response to artificial matrix landscapes of
A subset of these domains, such different dynamics and internalization frequencies diverse topology. These landscapes are generally engi-
as those found in the proteins when cells are plated on uniform substrates of varying neered to house symmetric or asymmetric microstruc-
zyxin, paxillin and testin, bind
actin filaments in a mechanical
compliance112–114. However, differences in endocytic tures or nanostructures of the synthetic substrates that
stress-​dependent manner. structures across single cells plated on stiffness gradi- are arrayed on a flat surface in specific patterns127–129
ents have not been shown. In addition, stretch-​activated or 3D channels of open or closed ratchets to mimic
LINC complex ion channels, such as PIEZO1/2, have been reported to topological features of tissue architecture in vivo130.
Linker of nucleoskeleton and
sense substrate stiffness115. One possibility is that as cells However, studies aimed at teasing apart the mechanisms
cytoskeleton (LINC) complex is
a complex of nuclear envelope apply traction force to the substrate on stiffness gradients, of landscape sensing to the downstream intra­cellular
proteins that connects the these channels will become differentially activated and events, which translate into motility, are sparse. In a
cytoskeleton to the nuclear produce local differences in migration-​relevant signal- few studies, cells were reported to sense topographical
lamina and is thus involved ling intermediates such as intracellular calcium. More features at nanometre to micrometre scales by extend-
in transferring signals from
sensing mechanical cues at
work will be required to test these ideas. ing leading-​edge protrusions. For instance, lamellipo-
the cell surface or in the Moving just inside the plasma membrane, many dia in fibroblasts were shown to orient themselves
cytosol into nucleus. cytoplasmic components of integrin-​containing focal parallel to the micropatterned lines coated with ECM

536 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

BAR-​family proteins
component, promoting elongated cell shape and direc- envelope triggered a contractile response that allowed
Bin/amphiphysin/Rvs161 tional migration131. In addition to parallel orientation, a cells to resist the physical compression and squeeze out
domain (BAR) proteins are smaller size of the protrusions has been reported to pro- of the confined space. Such a specific adaptive response
membrane-​binding proteins mote directed migration, possibly by limiting migration tailored by the nucleus may be of particular importance
that aid in regulating
perpendicular to the direction of ECM patterns131. In for immune cell patrolling through dense tissues, for
membrane shape.
contrast to lamellipodia used by fibroblasts, neurons use progenitor cells migrating through a densely packed
Arp2/3 complex both transient, non-​aligned and stabler, aligned filopo- cell mass during embryonic development or even during
A seven-​subunit protein dium populations to sense nanotopographical cues, and cancer cell invasion.
complex that possesses actin
to coordinate the signals that promote neurite outgrowth
nucleation and branching
activities leading to the
along ECM-​coated lined patterns132. In addition to the Galvanotaxis. How cells sense electric fields has been
generation of branched orientation of the structures, the availability of continu- debated for several decades138. Unlike mechanical cues,
actin networks. ous adhesive surfaces on the ECM-​coated aligned fibres which can be sensed either at the cell surface or inside
assists individual protrusions in sensing the topology133. the cell, electric fields must be sensed outside the plasma
N-​WASP
Such adhesion points give rise to the formation of membrane due to its high electrical resistance. The two
Neuronal Wiskott–Aldrich
syndrome protein activates the sequential focal adhesions along the continuous stretch predominant models for this sensing are membrane
Arp2/3 complex and promotes of fibres, which promote persistence of the protrusions depolarization and electromigration of surface proteins.
branched actin filament in a parallel orientation with respect to the aligned fibres. Recent reports favour electromigration of surface pro-
formation.
Local deformation or curvature of the plasma mem- teins as a mechanism of galvanotactic sensing139,140. In
Cortactin
brane at the interface of the cell and the substrate was these studies, the charge on the extracellular domains of
A nucleation promoting factor recently identified as the sensor of nanotopography134. model proteins, as well as some lipids and carbohydrates,
that activates the Arp2/3 Topography-​induced membrane curvature serves as a resulting from the application of an electric field induces
complex and promotes bridge between surface topography and intracellular electrophoresis of these molecules within the plane
branched actin filament
actin reorganization. Key pillars of the bridge include of the plasma membrane, either towards or away from
formation.
curvature-​s ensitive BAR-​family proteins, particularly the cathode140,141 (Fig. 2a). Moreover, it has been shown
FBP17, which recognize and accumulate within high-​ that galvanotaxis itself is sensitive to extracellular pH,
curvature areas present on either end of nanobars fab- which likely changes the charge state of proteins through
ricated to provide nanoscale topological cues. Localized protonation139.
FBP17 then facilitates nucleation of F-​actin by acti-
vating key actin cytoskeleton modulators, including Transmitting the signal
the Arp2/3 complex, N-​WASP and cortactin134. The result- In the third pillar of directed migration, the signal must
ing branched F-​actin network accumulates at both ends be transmitted from the sensor to the machinery nec-
of the nanobars and undergoes rapid polymerization– essary to move the cell. In some cases, this occurs via
depolymerization cycles. Of note, curvature sensing by polarized second messenger pathways, but in other
FBP17 recruitment is restricted to a curvature diam- cases the signal transduction machinery and the motil-
eter of less than 400 nm, indicating that topography ity machinery may overlap, such as during durotaxis.
sensing by curvature-​sensitive proteins is size specific. During the transmission step, a weak signal (in the form
Mechanistically, the identification of membrane curva- of a shallow gradient of the cue) is often amplified to
ture as the topology sensor fills the gap in our under- produce a robust cellular response.
standing of how topological cues from the surface
translate into actin fibre reorganization inside the cells Chemotaxis. For cells to migrate, their cytoskeletal
and, hence, is groundbreaking. machinery must be polarized142. The important ques-
Finally, a rather non-​conventional sensor of topo­ tions are how shallow gradients of extracellular chemi-
graphy is the cell nucleus. The nucleus is a large, bulky and cal cues can be transformed into steep polarized cellular
relatively rigid cellular organelle, repositioning of which responses, and at what point in the signalling cascade
is rate limiting during migration in constrained envi- are responses confined to a defined subcellular loca-
ronments. Recent studies have revealed that topological tion. The introduction of GFP technology and live-​cell
cues trigger changes in nuclear subcellular location and imaging have been critical in addressing this question.
shape, which have a significant impact on path finding It was established that chemotactic GPCRs remain uni-
and cell migration71,135,136. In the case of immune cells formly distributed in D. discoideum cells and neutrophils
using amoeboid migration, cytoskeletal forces position undergoing chemotaxis143,144, suggesting that the signals
the nucleus in the front portion of the cells. This allows leading to the segregation of the cytoskeletal machin-
cells to sample densely packed tissue, gauge the available ery are downstream of receptor occupancy. However, in
space and choose the path of least resistance71. Notably, lymphocytes, chemokine receptors have been reported
two recent studies proposed that the nucleus acts as to be clustered at the front of cells145. In mesenchymal
an internal ruler that interprets cell shape in confined tumour cells undergoing chemotaxis, the EGF receptor,
spaces, and facilitates rapid movement136,137. Specifically, like GPCRs, is also distributed homogeneously on the
it was shown that when the level of confinement is plasma membrane, but accumulates in endocytic vesicles
increased above a certain point sensed via nuclear on the side of the cell exposed to the high concentration
deformation, cells exert an active contractile force. This of the chemoattractant146. Because the EGF receptor sig-
contractile response was connected to the progressive nalling can continue from internalized endosomes, this
expansion and unfolding of the nuclear envelope with suggests that polarized receptor signalling could occur
increased confinement. A completely unfolded nuclear for RTKs in mesenchymal chemotaxis147.

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 537

0123456789();:
Reviews

a Polarization of intracellular b Signal transmission Signal sensing by receptor


signalling molecules
Signal transmission
Shallow gradient of directional cues RhoA PI3K TORC2 MAPK PLCγ

ROCK activation PtdIns(3,4,5)P3 DAG enriched at front


Rear Front enriched at front

PH domain-containing
MLCK proteins PKCα activation
Steep gradient of polarized
intracellular signalling molecules Rho-family GTPases
Myosin II light chain Myosin II inhibition
phosphorylation Arp2/3 complex at front

• Actin polymerization • Actomyosin contractility


• Front protrusion • Asymmetric force generation

c Signalling pathways triggered


by haptotactic cues
Fibronectin
(ECM)
Haptotaxis Chemoattractant Integrin
receptor cluster
Integrins
P
WAVE, Kindlin FAK P
WASP NPFs Src P
Arp2/3
Actin
ECM protein P
Chemoattractant
Collagen
fibre Rac Rho GEFs

Fig. 3 | Transmitting the signal. a | Cartoon depicting how shallow gradients of extracellular directional cues are
transmitted into steep gradients of intracellular signalling molecules at the front and rear of cells. b | Flow chart describing
how various signalling pathways activated by sensing of directional cues lead to changes in the cytoskeletal machinery.
c | Cartoon highlighting the molecular machinery that transmits haptotactic signals. Sensing gradients of fibronectin
through integrin engagement activates non-​receptor tyrosine kinases: focal adhesion kinase (FAK) and Src-​family
kinases. These kinases phosphorylate a variety of substrates, triggering the formation of new protein complexes, including
guanine nucleotide exchange factors (GEFs), and the activation of Rac, which leads to branched actin network formation
through Arp2/3 complex activation via the nucleation-​promoting factors (NPFs) WASP and WAVE, ultimately generating
lamellipodial structures. DAG, diacylglycerol; PH, pleckstrin homology; PI3K, phosphoinositide 3-​kinase; PtdIns(3,4,5)P3,
phosphatidylinositol 3,4,5-​trisphosphate.

Nevertheless, from findings in amoeboid cells it is that regulate actin assembly through the regulators of
clear that the polarization of signalling molecules at the Rho-​family GTPases, such as Rho guanine nucleotide
front and rear of cells undergoing chemotaxis occurs exchange factors, and DOCK–ELMO 153–155. However,
downstream of sensing receptors and upstream of the pharmacological inhibition or genetic ablation of PI3K
cytoskeletal machinery (Fig. 3a,b) (although the flow of does not completely inhibit chemotaxis, particularly in
actin may also contribute to the polarization of signal- steep chemotactic gradients152,156,157, and several parallel
Pleckstrin homology (PH)
ling molecules and hence cytoskeletal rearrangements pathways have been reported to regulate D. discoideum
domain may also fine-​tune the distribution and thus the recep- and neutrophil chemotaxis, including TORC2 (refs158–161),
Small protein domains of tion of the signal)148. Indeed, with use of probes that phospholipase A 2 (refs 162–166) and MAPK/ERK 167–170. In
approximately 120 amino specifically label lipids downstream of phosphoinos- fibroblasts undergoing chemotaxis towards the RTK
acids that are known to have
itide 3-​kinase (PI3K), it was shown that phosphatidy- ligand PDGF, the phospholipase PLCγ appears to be
phosphoinositide-​binding
specificity. linositol 3,4,5-​trisphosphate lipids are spatially restricted crucial for this process through localized hydrolysis of
to the leading edge of D. discoideum, neutrophils and phosphatidylinositol 4,5-​bisphosphate to yield diacyl­
DOCK–ELMO fibroblasts undergoing chemotaxis149–151. These polar- glycerol at the leading edge facing the highest concentra-
A protein complex consisting ized phosphatidylinositol 3,4,5-​t risphosphate sites, tion of PDGF156. This stable enrichment of diacylglycerol
of an adaptor protein, ELMO,
and a Rac-​specific guanine
which are dependent on Ras signalling (reviewed triggers the localized activation of the kinase PKCα
nucleotide exchange factor, in152), are then poised to spatially recruit a subset of and the subsequent inactivation or inhibition of myosin II
DOCK. pleckstrin homology (PH) domain-​containing proteins through non-​canonical phosphorylation of Ser1/Ser2

538 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

on the regulatory light chain, thereby creating asymme- considerably, and thus is not nearly as clear-​cut as dur-
try in myosin II-​mediated contractility. Furthermore, ing other directed migration types, such as chemotaxis.
in carcinoma cells migrating towards the RTK ligand However, the signal transduction mechanisms involved
EGF, amplification of the signal has been reported to during durotaxis can be broadly grouped into two
occur through the activation of the actin-​s evering classes, depending on the cell type and environmental
protein cofilin by PLCγ thereby allowing asymmetric context.
actin polymerization required for protrusion towards First, mechanically sensitive enzymes and proteins
the EGF gradient171. Finally, positive-​feedback and can generate traditional second messengers such as
negative-​feedback mechanisms centred at the regu- intracellular calcium changes. The best studied examples
lation of signal transduction via the Ras–PI3K–ERK of these are the non-​selective PIEZO1/2 stretch-​activated
pathway have been reported to be involved in reg- ion channels175. These proteins have been linked to a
ulating the modes of migration in D. discoideum172 wide variety of mechanosensation events but, to our
and the metastatic potential of epithelial cells173. Such knowledge, have not yet been specifically tested during
self-​organizing excitable signal transduction activities durotaxis. Cytoplasmic enzyme kinetics can also be con-
were proposed to underlie the control of the extension trolled by mechanical load176, which could theoretically
of actomyosin-​based protrusions and were suggested to lead to altered second messenger signalling, but this has
have a key role during development (reviewed in142). not been documented during mechanically triggered cell
Importantly, although many aspects of chemotactic migration.
signalling are common in amoeboid and mesenchymal Second, mechanical force can result in protein con-
cells, the fundamental difference in migration behav- formational changes, such as unfolding, or changes in
iours between these two types of cells, with specific biophysical properties, such as catch-​bond behaviour,
mechanisms, timescales and dynamics, requires distinct where mechanical load increases bond strength. One of
signalling pathways that are not fully understood92,93. the first proteins shown to display a mechanically sen-
sitive conformational change and altered binding part-
Haptotaxis. The transmission of haptotactic signals ners was p130Cas, an adaptor protein regulating tyrosine
and the transmission of chemotactic signals are similar, kinase-​based signalling related to cell adhesion118. This
involving traditional Rho-​family GTPase signalling path- paradigm of molecular stretching under mechani-
ways. The most well-​studied form of haptotaxis is sensing cal load has strongly influenced how we think about
gradients of ECM proteins through integrin engagement mechanosensing177. This has been further reinforced by
(Fig. 3c). In this case, integrin engagement leads to the the generation of Förster resonance energy transfer-​based
activation of the non-​receptor tyrosine kinases FAK molecular tension biosensors, including those of integrin
and Src-​family kinases174. These kinases phosphorylate adhesion-​associated vinculin and talin, which enabled
a variety of substrates, triggering the formation of new the visualization of local changes in tension that may
protein complexes including those involving guanine constitute signals for directed migration117,178–181. Both
nucleotide exchange factors, such as β-​PIX and TIAM1, vinculin and talin show F-​actin binding that is increased
and the activation of Rac, which leads to the formation of under mechanical load182,183. This catch-​bond binding for
lamellipodial structures. When these lamellipodia pro- both proteins is asymmetric, with a bias towards binding
trude up the gradient of fibronectin and encounter more the pointed (minus) end of the actin filament. In nascent
TORC2 ECM ligands for the integrins, a positive-​feedback loop is adhesion structures at the leading edge, the proximal
Target of rapamycin complex 2 established that is critical for haptotaxis of fibroblasts on actin networks — which generally face the membrane
is composed of seven conserved gradients of fibronectin. However, it is not clear whether with their barbed (plus) ends — are flowing backwards
subunits and is involved in
regulating proliferation,
signal amplification akin to what happens during chemo­ by retrograde flow from the leading edge. This polar-
survival, cell migration and taxis occurs or is required during ECM haptotactic ized flowing population of filaments can be engaged by
cytoskeletal reorganization. signalling. In the case of haptotactic migration on gra- vinculin and talin associated with the nascent adhesions
dients of surface-​bound chemokines, the presumption due to their asymmetric catch-​bond properties, possibly
Phospholipase A2
is that the signalling pathways activated by the cognate contributing to localized adhesion maturation and hence
An enzyme that cleaves
phospholipids to give rise GPCRs are the same as the signalling pathways activated directional migration. The next step will be to observe
to lipid products (arachidonic by the diffusible version of the chemotactic cue, but this how these local, tension-​dependent conformational
acid or lysophosphatidic acid) presumption needs further experimental validation. changes translate to more global mechanotransduction
that either have the ability Interestingly, one study reported differential regulation networks across single cells migrating on gradients of
to regulate signalling events
or are substrates in the
of CCR7 towards haptotactic ECM-​bound gradients of substrate stiffness.
generation of bioactive lipids. CCL21 versus chemotactic, diffusible CCL21 (ref.103).
This suggests that a cell response to haptotactic versus Topotaxis. How guidance signals from tissue topogra-
MAPK/ERK chemo­tactic cues, even when involving the same receptor/ phy are transmitted within cells is not clearly defined.
A group of protein kinases
pathway, may be specifically modulated to adjust cell One can speculate that it will involve calcium, in light
that transduce signals from
cell surface receptors to behaviour to the migratory context. of the well-​recognized role of calcium in converting
the nucleus. external information into biological signals that subse-
Durotaxis. Delineating how durotactic signals are quently lead to actin polymerization. Enhanced intra-
Förster resonance energy transmitted is challenging as the ‘receptor’ or ‘recep- cellular calcium activity has been detected in astrocytes
transfer
A mechanism describing
tors’ responding to durotactic cues are not definitively seeded onto a micropatterned surface184. Astrocytes were
energy transfer between two known. Furthermore, the distinction between sensing found to be elongated and aligned along the direction of
light-​sensitive molecules. and transmitting a mechanical signal could overlap the grooves accompanied by frequent calcium peaks in the

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 539

0123456789();:
Reviews

aligned cells relative to randomly oriented, rounded Consistent with this finding, galvanotaxis is sensitive
cells on flat surfaces184. Whether such calcium activity to many pharmacological inhibitors and genetic per­
also tunes cell motility on such patterned surfaces is less turbation of intracellular signalling pathways 77,188.
clear. It has been partly addressed using surface topol- In this way, galvanotaxis is most similar to chemot-
ogy with different degrees of confinement that mimic axis in terms of signal transmission. Consistent with
narrow channels and fibre-​like tracks of natural ECM this idea, a large-​scale genetic screen for D. discoideum
in vivo. In one of the recent studies, elevated intracellu- mutants defective in galvanotaxis revealed an impres-
lar calcium concentration was found to be important in sive overlap of identified genes with those previously
mobilizing cells through confined microchannels, where implicated in chemotaxis188. The activation of a wide
the confinement-​driven force was transmitted inside the array of signalling pathways may arise due to the
cells as calcium influx via activation of PIEZO1 (ref.185). electromigration-​based clustering of a range of cell sur-
However, cell migration in the study was induced in face receptors on either side of the cell (relative to the
the presence of a chemotactic source, indicating the electric field) that somehow activates downstream, intra-
need to further investigate whether topology or con- cellular signalling pathways leading to polarized signal
fined space-​driven calcium influx controls cell motility transduction139,140. Indeed, recent data indicate that local-
independently of chemical cues. ized, photo-​induced clustering of GPCRs can induce
Furthermore, PIEZO1 activation was recently shown ligand-​independent signalling189. Interestingly, in kerato­
to be regulated by the geometric features, such as rough- cytes, inhibition of one of the kinases commonly asso-
ness and stiffness, of the surrounding substrate when ciated with galvanotaxis, PI3K, can actually reverse
force was applied externally by substrate deflection at the polarity of galvanotaxis, leading to migration of
cell–substrate interfaces186. For instance, when indi- cells towards the anode rather than the cathode139.
vidual pillars of a deformable micropillar array were Although the mechanism of this reversal is not known,
deflected, the amplitude of the PIEZO1-​mediated cur- it may indicate that electric field-​induced signalling is
rent was higher in cells surrounded by sparsely arrayed being activated on both the anode-​facing side and the
pillars, which led to less substrate roughness than for cathode-​facing side of the cell, resulting in a ‘tug of war’
the densely arrayed pillars. Notably, the amplitude of the for cell polarization decisions that can be influenced by
PIEZO1-​mediated current induced by pillar deflection perturbation of intracellular signalling.
decreased when cells contacted substrates with greater
stiffness but relatively low roughness186. Whether mod- Executing the signal
ulation of PIEZO1 channel activation by substrate The translocation of cells, individually or in groups,
mechanics affects calcium spatio-​temporal dynamics relative to their environment requires the generation of
and ultimately cell migration in response to various asymmetric or polarized forces relative to the substrate.
degrees of compressive, tensile and shear forces relevant There are several sources of this force generation, but a
in physiological contexts, such as dense tissues or blood key concept is that directed migration-​promoting sig-
capillaries, remains to be addressed. nalling biases these forces either towards or away from
There is also evidence that topotactic cues, and in environmental cues. Directed cell migration cues do not
particular cell compaction, are sensed via the nucleus, activate special mechanisms of cell translocation but bias
which will inevitably undergo deformation in this con- the migration and cell polarity machinery that operates
text, and that both calcium and the lipid-​based second during normal, random cell migration12. Depending
messenger arachidonic acid are essential to transmit this on the mode of migration used by a particular cell type
signal to the cytoskeleton71,136,137. In this mechanism, (Box 1), multiple directed migration cues can converge
nuclear envelope stretching and unfolding resulting from on a common set of force generation mechanisms lead-
the compressive force of compaction is associated with ing to cell translocation. Thus, with this fourth pillar of
calcium release from internal stores, possibly through directed migration, we will not separate the directed
stretch-​sensitive calcium channels. This then triggers migration forms by the environmental cue; rather, we
Pseudopodia
the redistribution of the calcium-​dependent phospho- will discuss the different forms of asymmetric force gen-
Protrusive structures in
amoeboid cells generated lipase A2 (cPLA2) from the cytosol to the stretched eration activated during directed migration (Fig. 4) and
by branched and linear actin nuclear envelope. Subsequent activation of cPLA2 leads describe how the signal transmission step regulates this
filament arrays in the leading to release of arachidonic acid from the nuclear envelope force generation.
edge and aligned with the phospholipids. cPLA2-​mediated release of arachidonic
direction of movement.
acid is critical for the biosynthesis of lipid mediators, Protrusion-​based mechanisms. One of the character-
Ena/VASP including LTB4, which have well-​established roles in istic features of most migrating cells is that they have
Enabled/vasodilator-​stimulated activating myosin contractility and immune cell migra- some kind of protrusive structure at their leading edge,
phosphoproteins are actin tion in an autocrine and paracrine manner42,187. Notably, which is aligned with the direction of movement (Fig. 4a).
polymerases that drive actin
LTB4-​synthesizing enzymes also reside in the nucleus, With a few notable exceptions, such as nematode sperm
filament elongation and
antagonize filament capping, which thereby identifies the nucleus as a dynamic hub for cells190, most protrusions are driven by the polymeriza-
leading to the generation both sensing elements and second messenger production tion of actin filament arrays191. In amoeboid cells, these
of linear actin filaments. during cell motility in response to confined topology. are known as pseudopodia, and the actin is a mixture
of branched actin networks produced by the activation of
Formin
A group of actin polymerases
Galvanotaxis. Cells in electric fields activate a wide the Arp2/3 complex and linear actin arrays produced
that drives the formation variety of intracellular signalling pathways, including by Ena/VASP and formin proteins192. In fibroblasts and
of linear actin filaments. AKT, Src-​family kinases, MEK–ERK and JAK1 (ref.77). other more adherent cells, the protrusions often take on

540 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

a Protrusion-based mechanisms Branched b Contractility-based mechanisms


actin network Stress fibre based
Contractile
Actin Actomyosin force
network forming Actomyosin
Pseudopod stress fibre bundle

Actomyosin Myosin Myosin


network Integrin
Focal adhesion
complex
Collagen fibre
Lamellipodium
Blebbing based
Actin cortex
Filopodium Myosin Polarized New membrane–
Linker Asymmetric blebbing cortex attachments
protein membrane
delamination
Local from the cortex
Linear increases
actin filament in cortical
tension
Local rupture
of the cortex

c Alternative mechanisms
Nuclear piston based Friction based Molecular paddling based
Low pressure Actin cortex Transmembrane
zone at the back protein anchored
of the nucleus to the crotex

Back Front

High-pressure
Confined 3D space zone in front Cortex
of the nucleus retrograde
Cortex flow
retrograde flow Advection

Fig. 4 | Executing the signal. Depiction of how asymmetric force is generated through protrusion-​based, contractility-​
based or alternative mechanisms. a | Formation of protrusions such as pseudopodia, lamellipodia and/or filopodia is driven
by branched and linearly polymerized actin networks. b | Contractility-​based mechanisms of migration (characteristic
of mesenchymal cells) depend on the establishment of stress fibres, where a contractile array of actomyosin networks is
mechanically coupled to the substrate through integrin-​based focal adhesions (top). Many cell types, in particular in the
in vivo context, migrate via extension of blebs (bottom), which are generated through local increases in cortical tension
on the non-​blebbing side of the cell (marked with arrows) and asymmetric membrane tearing (delamination), or by local
rupture of the cortex, or both. c | Cells moving through a confined space use the nuclei as a piston to generate zones
of high cytosolic pressure at the leading edge. Cells moving in the absence of substrate adhesion depend on friction
between cells and the environment, generated by retrograde flow of the cortical actin. Cells can also use molecular
paddling to swim through the environment using horizontal rearward flow of transmembrane proteins anchored to the
actin cortex (advection).

a flatter appearance and fan-​like architecture (referred migration cues193,197. For example, one of the most sat-
to as ‘lamellipodia’) that is more dominated by Arp2/3-​ isfying connections between signalling pathways and
branched actin, although they also contain linear protrusions is the activation of the Arp2/3 complex, via
actin microspikes that can extend beyond the edge as Rho-​family GTPases and nucleation-​promoting factors
filopodia193. Some cells, such as neuronal growth cones, such as SCAR/WAVE, a topic that has been extensively
have protrusions that are almost entirely formed by reviewed elsewhere191,198. However, several groups have
filopodia, containing bundled linear arrays of actin with shown that cells lacking the Arp2/3 complex can still
concentrated actin polymerases such as Ena/VASP and undergo chemotaxis in various contexts, albeit with
formins at their tip194. Of note, these different protru- reduced migration efficiency197,199–201. This indicates
sions are not finite entities and are highly plastic and can that the Arp2/3-​branched actin pathway is not strictly
dynamically interconvert over time, such as when filopo- required for chemotaxis in all circumstances and points
dia direct where lamellipodia can form195 or when lamel- towards other possible mechanisms of asymmetric force
SCAR/WAVE lipodia yield clusters of filopodia196. The proportion of generation.
Suppressor of cAR/WASP actin network types within protrusions (branched versus Other mechanisms responsible for triggering or
family verprolin-​homologous linear) is probably more important than the historical tuning protrusions have been identified. In the case of
protein is a nucleation-​
promoting factor that activates
naming convention for the protrusion type. haptotaxis, the activation of the Arp2/3 complex and
actin nucleation activity of the Over the years, many lines of evidence have linked the subsequent generation of branched actin networks
Arp2/3 complex. the formation of protrusions with various directed at nascent integrin adhesions requires both FAK–Src

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 541

0123456789();:
Reviews

and the RAC1 GTPase, integrated through the WAVE cluster of cells was shown to have a mechanically con-
regulatory complex174,197 (Fig. 3c). In the case of neuronal tinuous band of actomyosin contractility around the
growth cones responding to chemotactic guidance cues, periphery of the cluster that can be locally inhibited by
emerging evidence suggests that monoubiquitinylation chemotactic cues at the front to produce a form of ‘rear
of VASP mediated by the ubiquitin ligase TRIM9 helps wheel’ drive motility213.
tune the activity of this actin polymerase at the tips of At the single-​cell level, localized control of contrac-
filopodia202. In the case of durotaxis, both Arp2/3-​based tility (either positive or negative) has a profound effect
lamellipodia203 and filopodia204 are required to sense and on polarization and directed migration214. In fibroblasts
respond to varying substrate stiffness, although whether migrating in a gradient of the chemoattractant PDGF,
these protrusions are regulated through traditional, sec- local inhibition of myosin II at the leading edge is criti­cal
ond messenger-​type signalling pathways that are respon- for chemotaxis156. At the opposite end of the cell, recent
sive to substrate stiffness remains to be determined. One studies in neutrophils and keratocytes have shown that
consistent mechanism of protrusion control is the split- control of rear contractility governs the direction of
ting of existing protrusions and the selective stabilization cell movement215,216. Even in cells that lack strong cell–
of the protrusion that is oriented more strongly towards substrate or cell–cell adhesions, such as amoeboid
the gradient of the signal. This has been demonstrated tumour cells, local contractility may have a key role in
in the chemotactic migration of both D. discoideum the generation of cellular blebs through local increases
and fibroblasts205,206, and may reflect a common strat- in cortical tension on the non-​blebbing side of the
egy for many forms of directed migration. Finally, in cell, local relaxation of the cortex to permit blebbing
cells undergoing topotaxis, including D. discoideum, or both217. However, the signals upstream of polarized
neutrophils and breast cancer cells, it was observed that blebbing are incompletely understood218.
asymmetric microstructures of the substratum induce
actin waves, which propagate unidirectionally, defining Alternative mechanisms via pistons, ridges and molecu-
the direction of cell translocation. Curiously, the waves lar paddles. Beyond actin-​based protrusions and acto-
could propagate in different directions, depending on myosin contractility, cells have other ways to generate
the cell type, and this was associated with the establish- asymmetric traction force relative to their environment
ment of different focal adhesion patterns and differences and move (Fig. 4c). However, most of these have not
in membrane dynamics and protrusion extension, which been definitively linked to specific directed migration
were linked to distinct local cell cortical plasticity in cues. Cells moving through confined environments
different cell types129,207. can use their nucleus as a piston to generate cytoplas-
mic pressure gradients at the leading edge to facilitate
Contractility-​based mechanisms. The other main gen- migration126. Possible cellular regulation of this ‘nuclear
erator of asymmetric force relative to the substrate for piston’ mode could involve the association of the nucleus
Calpain migrating cells is contractile arrays of actin and non-​ with lateral membrane or localized osmotic control.
Calcium-​activated cysteine muscle myosin II (ref.208) (Fig. 4b). In the case of firmly Other mechanisms are observed in immune cells,
protease that cleaves adhesion adherent cells, such as mesenchymal cells, these arrays which do not require integrins for their migration in 3D
complex proteins.
of contractile actin form stress fibres that are mecha­ environments111. In one recent study it was shown that
Border cell nically coupled to the substrate through integrin-​based T cells can translocate using friction generated between
A specialized cell type focal adhesions. Asymmetric strengthening or weak- the cell and its environment, which is coupled to cellu-
that migrates as a group ening of these adhesion structures provides a dynamic lar translocation through retrograde flow of the actin
through the egg chambers in way to control the direction of cell migration. Indeed, cortex219. In another recent study, non-​adherent leuko-
Drosophila melanogaster.
many signalling pathways have been linked to adhe- cytes were shown to ‘swim’ through their environment
Blebs sion turnover, including phosphorylation of specific using a fascinating ‘molecular paddling’ mechanism,
Spherical membrane adhesion proteins209 and the myosin II regulatory light which is mediated by transmembrane proteins coupled
protrusions that rely on myosin-​ chain156, selective proteolytic cleavage of adhesion pro- to the actin cortex and moving by advection across
based contraction and pressure-​
teins by calpain210 and relaxation of adhesions through the cell surface in a coherent manner relative to their
driven cytosolic flow. Bleb-​like
protrusions are commonly contacts with microtubules68. Remodelling of focal environment220. This is a remarkably efficient migration
used for motility by amoebas adhesions almost certainly underpins durotactic migra- strategy, with cell speeds similar to those of adhesion-​
and embryonic cells. However, tion in many cell types122,209 but may also be relevant for based migration. Such mechanisms could be of great
leukocytes and tumour cells chemotactic migration of fibroblasts156. importance for migration through in vivo environments,
can use blebbing motility
especially in 3D environments
In the case of cells with robust cell–cell adhesions, given that cellular environments are characterized by a
under confined conditions. such as sheets of epithelial cells, myosin-​based con- low Reynolds number221 — meaning that at cellular scales,
tractility can also be a potent mechanism to direct inertia and momentum are negligible and thus move-
Reynolds number migration211,212. In this case, the contractile arrays are ment requires strategies different from those for human-​
A dimensionless number
linked to the cell–cell adhesion sites and control the relevant length scales. However, it is unclear whether this
important in fluid mechanics.
Cellular scales are inherently shape of cells relative to their neighbours, as well as coor- paddling mechanism is used only for random migration
a low Reynolds number dinate the transmission of mechanical force across mul- or is also responsive to external environmental cues.
environment where inertia and tiple cells. Cells with strong intercellular junctions often
momentum are negligible move collectively as groups, as in the case of border cell Conclusions and perspective
and thus movement requires
strategies different from those
migration in D. melanogaster and neural crest migration in Directed migration has been studied for many decades,
for human-​relevant length vertebrates. In recent work on neural crest migration but our understanding of the mechanisms underpinning
scales. in zebrafish and Xenopus laevis embryos, the migrating this process has increased dramatically only over the past

542 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

Box 2 | Key open questions for specific modes of directed cell migration information. Multiple approaches to problems as com-
plex as directed migration are required to achieve the
Chemotaxis level of understanding necessary to manipulate this
How are stable gradients of diffusible cues generated and maintained in tissues with process or develop therapeutic interventions.
interstitial fluid flows as in most multicellular organisms?
A particularly difficult aspect of understanding
Haptotaxis directed migration in vivo is that cells are being exposed
Is haptotaxis using extracellular matrix as a migratory cue used only by mesenchymal to multiple migration-​inducing cues simultaneously.
cells that have strong substrate adhesion? Do low-​adhesion amoeboid cells still sense This ‘multicue’ problem is both fascinating and experi-
these cues? mentally challenging. For example, if a cell is presented
Durotaxis with a chemotactic cue orienting its migration in one
How do cells sense durotactic cues? Are there multiple/cell type-​specific ‘receptors’ direction but a topological or durotactic cue is orienting
for durotaxis, and how do they connect to the cell polarity and translocation machinery? it in the opposite direction, how does the cell prioritize
Topotaxis or integrate these conflicting signals? One compelling
How are topotactic cues sensed by cells? Are there different mechanisms that are recent example of this problem comes from D. melano­
specific to particular 2D or 3D geometries? How does topology sensing by membrane gaster border cell migration where the relative con-
curvature-​sensitive proteins affect leading-​edge protrusions and translate into guided tributions of chemotactic and topological cues were
migration? dissected by elegant genetic approaches223. However,
Galvanotaxis knowing the specific perturbations necessary to dissect
Electrophoretic migration of cell surface receptors within the plane of the plasma multicue migration in vivo requires a specific under-
membrane and their activation seems to be critical for galvanotaxis, but how are those standing of the underlying processes, and more work in
receptors activated by electric current-​induced clustering? Is there polarized autocrine simplified, single-​cue systems will be needed. In a related
secretion of ligands that accompanies this clustering or is clustering of receptors problem, how do cells switch their migration direction?
sufficient to activate signalling in a ligand-​independent manner?
For example, neutrophils initially home to injured sites
but later reverse their migration and move away from
the inflamed area as part of the resolution process224–226.
10 years. New approaches for producing stable and/or Again, a more comprehensive understanding of the
tunable gradients of various factors, controlled substrate mechanisms of directed migration will be required to
geometries and new methods for imaging cells in 3D, understand complex cases such as these.
physiological contexts have expanded our knowledge of A deeper understanding of directed cell migration
the molecular and cellular processes required to execute will be necessary to effectively treat human diseases.
directed migration. The unifying, conceptual framework Interventions that could block tumour cells from leaving
presented herein has allowed us to highlight this pro- the primary tumour or intravasating into blood or lymph
gress. Going forwards, we hope that this framework vessels could be a potent way to block metastasis —
will prompt the field to fill in gaps in our understanding the clinically most deadly aspect of cancer. Similarly,
(see Box 2). attenuating the recruitment of immune cells to inflamed
One of the broader challenges in studying directed areas in the context of chronic illnesses such as arthritis,
cell migration is understanding it in physiological con- asthma and atherosclerosis could improve disease out-
texts. Studying cell migration on the rigid, 2D surfaces come. Conversely, encouraging the migration of subsets
commonly used for microscopy has been criticized of immune cells into tumours could improve antican-
as not reflecting these physiological contexts222. To be cer immune therapies. The inappropriate migration of
fair, the tools and imaging approaches needed to study immune cells or fibroblasts can also lead to autoimmune
migration in these more physiological contexts or in disorders and fibrosis, respectively. Finally, finding ways
living organisms have only recently become widely for neuronal cells to ignore ‘stop’ cues such as in nerve
available to researchers, and the field will need to con- injuries could lead to regenerative treatments for paraly­
tinue to embrace this transition. However, studying sis. Understanding how cells, using different migration
directed cell migration using simplified 2D systems modes (for example, amoeboid versus mesenchymal),
and model organisms has revealed underlying princi- migrate in response to the various chemical, mecha­
ples and insights that have become the building blocks nical, geometric and electrical cues that are present
to understand migration in more complex systems. By in these disease states has the potential to lead to the
analogy, studying the biochemical properties of purified identification of novel therapeutic avenues.
proteins, even though these systems do not replicate the
complexity of the cytoplasm, continues to yield essential Published online 14 May 2021

1. Borrell, V. Recent advances in understanding migration and invasion. Br. J. Cancer https://doi.org/ 8. Abercrombie, M., Dunn, G. A. & Heath, J. P.
neocortical development. F1000Res https://doi.org/ 10.1038/s41416-020-01149-0 (2020). The shape and movement of fibroblasts in culture.
10.12688/f1000research.20332.1 (2019). 5. García-​Cuesta, E. M. et al. The role of the CXCL12/ Soc. Gen. Physiol. Ser. 32, 57–70 (1977).
2. Pawar, K. B., Desai, S., Bhonde, R. R., Bhole, R. P. CXCR4/ACKR3 axis in autoimmune diseases. 9. Sardelli, L. et al. Engineering biological gradients.
& Deshmukh, A. A. Wound with diabetes: Front. Endocrinol. 10, 585 (2019). J. Appl. Biomater. Funct. Mater. 17,
present scenario and future. Curr. Diabetes Rev. 6. Glass, D. S. et al. Idiopathic pulmonary fibrosis: 2280800019829023 (2019).
https://doi.org/10.2174/157339981666620070318 molecular mechanisms and potential treatment 10. Buttenschön, A. & Edelstein-​Keshet, L. Bridging from
0137 (2020). approaches. Respir Investig. 58, 320–335 single to collective cell migration: a review of models
3. Janssen, E. & Geha, R. S. Primary immunodeficiencies (2020). and links to experiments. PLoS Comput. Biol. 16,
caused by mutations in actin regulatory proteins. 7. Abercrombie, M. The Croonian Lecture, e1008411 (2020).
Immunol. Rev. 287, 121–134 (2019). 1978 - the crawling movement of metazoan cells. 11. Wilkinson, P. C. Random locomotion; chemotaxis
4. Novikov, N. M., Zolotaryova, S. Y., Gautreau, A. M. Proc. Royal Soc. B Biol. Sci. 207, 129–147 and chemokinesis. a guide to terms defining cell
& Denisov, E. V. Mutational drivers of cancer cell (1980). locomotion. Immunol. Today 6, 273–278 (1985).

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 543

0123456789();:
Reviews

12. Petrie, R. J., Doyle, A. D. & Yamada, K. M. Random migration through competing mechanisms. Proc. Natl 62. Cai, L., Xiong, X., Kong, X. & Xie, J. The role of the lysyl
versus directionally persistent cell migration. Acad. Sci. USA 108, 11115–11120 (2011). oxidases in tissue repair and remodeling: a concise
Nat. Rev. Mol. Cell Biol. 10, 538–549 (2009). 37. Müller, P., Rogers, K. W., Yu, S. R., Brand, M. & review. Tissue Eng. Regen. Med. 14, 15–30 (2017).
13. Dorward, D. A. et al. The role of formylated peptides Schier, A. F. Morphogen transport. Development 140, 63. Handorf, A. M., Zhou, Y., Halanski, M. A. & Li, W. J.
and formyl peptide receptor 1 in governing neutrophil 1621–1638 (2013). Tissue stiffness dictates development, homeostasis,
function during acute inflammation. Am. J. Pathol. 38. Restrepo, S., Zartman, J. J. & Basler, K. Coordination and disease progression. Organogenesis 11, 1–15
185, 1172–1184 (2015). of patterning and growth by the morphogen DPP. (2015).
14. Ehrengruber, M. U., Geiser, T. & Deranleau, D. A. Curr. Biol. 24, R245–R255 (2014). 64. Ye, M. et al. Evolving roles of lysyl oxidase family in
Activation of human neutrophils by C3a and C5A. 39. Kriebel, P. W., Barr, V. A., Rericha, E. C., Zhang, G. tumorigenesis and cancer therapy. Pharmacol. Ther.
Comparison of the effects on shape changes, & Parent, C. A. Collective cell migration requires 215, 107633 (2020).
chemotaxis, secretion, and respiratory burst. vesicular trafficking for chemoattractant delivery at 65. Alcudia, J. F. et al. Lysyl oxidase and endothelial
FEBS Lett. 346, 181–184 (1994). the trailing edge. J. Cell Biol. 183, 949–961 (2008). dysfunction: mechanisms of lysyl oxidase down-​
15. Sadik, C. D. & Luster, A. D. Lipid-​cytokine-chemokine 40. Kriebel, P. W. et al. Extracellular vesicles direct regulation by pro-​inflammatory cytokines.
cascades orchestrate leukocyte recruitment in migration by synthesizing and releasing chemotactic Front. Biosci. 13, 2721–2727 (2008).
inflammation. J. Leukoc. Biol. 91, 207–215 (2012). signals. J. Cell Biol. 217, 2891–2910 (2018). 66. Chen, Y., Peng, W., Raffetto, J. D. & Khalil, R. A.
16. McDonald, B. & Kubes, P. Cellular and molecular 41. Esser, J. et al. Exosomes from human macrophages Matrix metalloproteinases in remodeling of lower
choreography of neutrophil recruitment to sites of and dendritic cells contain enzymes for leukotriene extremity veins and chronic venous disease.
sterile inflammation. J. Mol. Med. 89, 1079–1088 biosynthesis and promote granulocyte migration. Prog. Mol. Biol. Transl Sci. 147, 267–299 (2017).
(2011). J. Allergy Clin. Immunol. 126, 1032–1040 (2010). 67. Quintero-​Fabián, S. et al. Role of matrix
17. Junger, W. G. Immune cell regulation by autocrine 42. Afonso, P. V. et al. LTB4 is a signal-​relay molecule metalloproteinases in angiogenesis and cancer.
purinergic signalling. Nat. Rev. Immunol. 11, during neutrophil chemotaxis. Dev. Cell 22, Front. Oncol. 9, 1370 (2019).
201–212 (2011). 1079–1091 (2012). 68. Kaverina, I., Krylyshkina, O. & Small, J. V. Microtubule
18. Yoo, S. K., Starnes, T. W., Deng, Q. & Huttenlocher, A. 43. Lämmermann, T. et al. Neutrophil swarms require targeting of substrate contacts promotes their
Lyn is a redox sensor that mediates leukocyte wound LTB4 and integrins at sites of cell death in vivo. Nature relaxation and dissociation. J. Cell. Biol. 146,
attraction in vivo. Nature 480, 109–112 (2011). 498, 371–375 (2013). 1033–1044 (1999).
19. Katikaneni, A. et al. Lipid peroxidation regulates 44. Brown, M. et al. Lymphatic exosomes promote 69. Paul, C. D., Mistriotis, P. & Konstantopoulos, K.
long-​range wound detection through 5-lipoxygenase dendritic cell migration along guidance cues. Cancer cell motility: lessons from migration in confined
in zebrafish. Nat. Cell Biol. 22, 1049–1055 (2020). J. Cell. Biol. 217, 2205–2221 (2018). spaces. Nat. Rev. Cancer 17, 131–140 (2017).
20. Blockus, H. & Chédotal, A. Slit-​Robo signaling. 45. Chen, T., Guo, J., Yang, M., Zhu, X. & Cao, X. 70. Wolf, K., Müller, R., Borgmann, S., Bröcker, E. B.
Development 143, 3037–3044 (2016). Chemokine-​containing exosomes are released from & Friedl, P. Amoeboid shape change and contact
21. Boyer, N. P. & Gupton, S. L. Revisiting netrin-1: one heat-​stressed tumor cells via lipid raft-​dependent guidance: T-​lymphocyte crawling through fibrillar
who guides (axons). Front. Cell Neurosci. 12, 221 pathway and act as efficient tumor vaccine. collagen is independent of matrix remodeling by
(2018). J. Immunol. 186, 2219–2228 (2011). MMPs and other proteases. Blood 102, 3262–3269
22. Hu, S. & Zhu, L. Semaphorins and their receptors: 46. Lim, K. et al. Neutrophil trails guide influenza-​specific (2003).
from axonal guidance to atherosclerosis. Front. CD8+ T cells in the airways. Science 349, aaa4352 71. Renkawitz, J. et al. Nuclear positioning facilitates
Physiol. 9, 1236 (2018). (2015). amoeboid migration along the path of least resistance.
23. Kania, A. & Klein, R. Mechanisms of ephrin-​Eph 47. Sung, B. H., Ketova, T., Hoshino, D., Zijlstra, A. & Nature 568, 546–550 (2019).
signalling in development, physiology and disease. Weaver, A. M. Directional cell movement through This seminal article shows that leukocytes position
Nat. Rev. Mol. Cell Biol. 17, 240–256 (2016). tissues is controlled by exosome secretion. the nucleus frontwards to gauge the available
24. Crick, F. Diffusion in embryogenesis. Nature 225, Nat. Commun. 6, 7164 (2015). space and choose their path.
420–422 (1970). 48. Sung, B. H. & Weaver, A. M. Exosome secretion 72. Castro-​Castro, A. et al. Cellular and molecular
25. Majumdar, R., Sixt, M. & Parent, C. A. New paradigms promotes chemotaxis of cancer cells. Cell Adh. Migr. mechanisms of MT1-MMP-​dependent cancer cell
in the establishment and maintenance of gradients 11, 187–195 (2017). invasion. Annu. Rev. Cell Dev. Biol. 32, 555–576
during directed cell migration. Curr. Opin. Cell Biol. 49. Hynes, R. O. Stretching the boundaries of extracellular (2016).
30, 33–40 (2014). matrix research. Nat. Rev. Mol. Cell Biol. 15, 73. Wyckoff, J. B., Pinner, S. E., Gschmeissner, S.,
26. Lau, S. et al. A negative-​feedback loop maintains 761–763 (2014). Condeelis, J. S. & Sahai, E. ROCK- and myosin-​
optimal chemokine concentrations for directional cell 50. Charras, G. & Sahai, E. Physical influences of the dependent matrix deformation enables protease-​
migration. Nat. Cell Biol. 22, 266–273 (2020). extracellular environment on cell migration. independent tumor-​cell invasion in vivo. Curr. Biol. 16,
27. Stapornwongkul, K. S., de Gennes, M., Cocconi, L., Nat. Rev. Mol. Cell Biol. 15, 813–824 (2014). 1515–1523 (2006).
Salbreux, G. & Vincent, J. P. Patterning and growth 51. Mouw, J. K., Ou, G. & Weaver, V. M. Extracellular 74. Fisher, K. E. et al. MT1-MMP- and Cdc42-dependent
control in vivo by an engineered GFP gradient. Science matrix assembly: a multiscale deconstruction. signaling co-​regulate cell invasion and tunnel
370, 321–327 (2020). Nat. Rev. Mol. Cell Biol. 15, 771–785 (2014). formation in 3D collagen matrices. J. Cell Sci. 122,
Using a synthetic morphogen in D. melanogaster, 52. Bonnans, C., Chou, J. & Werb, Z. Remodelling the 4558–4569 (2009).
the authors show that the expression of non-​ extracellular matrix in development and disease. 75. Weigelin, B., Bakker, G. J. & Friedl, P. Intravital
signalling decoy receptors improves the ability of Nat. Rev. Mol. Cell Biol. 15, 786–801 (2014). third harmonic generation microscopy of collective
receptors engineered to respond to the synthetic 53. Albacete-​Albacete, L. et al. ECM deposition is driven melanoma cell invasion: Principles of interface
morphogen to organize patterning and growth by caveolin-1-dependent regulation of exosomal guidance and microvesicle dynamics. Intravital 1,
in vivo. biogenesis and cargo sorting. J. Cell Biol. https://doi. 32–43 (2012).
28. Tweedy, L., Knecht, D. A., Mackay, G. M. & Insall, R. H. org/10.1083/jcb.202006178 (2020). 76. Du Bois-​Reymond, E. H. Vorläufiger Abriss einer
Self-​generated chemoattractant gradients: attractant 54. Zimmerman, S. P., Asokan, S. B., Kuhlman, B. & Untersuchung über den sogenannten Froschstrom und
depletion extends the range and robustness of Bear, J. E. Cells lay their own tracks - optogenetic über die elektromotorischen Fische. Ann. Phys. https://
chemotaxis. PLoS Biol. 14, e1002404 (2016). Cdc42 activation stimulates fibronectin deposition doi.org/10.1002/andp.18431340102 (1843).
29. Muinonen-​Martin, A. J. et al. Melanoma cells break supporting directed migration. J. Cell Sci. 130, 77. Zhao, M. et al. Electrical signals control wound healing
down LPA to establish local gradients that drive 2971–2983 (2017). through phosphatidylinositol-3-OH kinase-​gamma and
chemotactic dispersal. PLoS Biol. 12, e1001966 55. Singh, P., Carraher, C. & Schwarzbauer, J. E. Assembly PTEN. Nature 442, 457–460 (2006).
(2014). of fibronectin extracellular matrix. Annu. Rev. Cell Dev. 78. Hotary, K. B. & Robinson, K. R. Endogenous electrical
30. Juin, A. et al. N-​WASP control of LPAR1 trafficking Biol. 26, 397–419 (2010). currents and voltage gradients in Xenopus embryos
establishes response to self-​generated LPA gradients 56. Miller, C. G., Budoff, G., Prenner, J. L. & and the consequences of their disruption. Dev. Biol.
to promote pancreatic cancer cell metastasis. Dev. Cell Schwarzbauer, J. E. Minireview: fibronectin in retinal 166, 789–800 (1994).
https://doi.org/10.1016/j.devcel.2019.09.018 (2019). disease. Exp. Biol. Med. 242, 1–7 (2017). 79. Klein, P. S. et al. A chemoattractant receptor controls
31. Tweedy, L. et al. Seeing around corners: cells solve 57. Schultz, G. S. & Wysocki, A. Interactions between development in Dictyostelium discoideum. Science
mazes and respond at a distance using attractant extracellular matrix and growth factors in wound 241, 1467–1472 (1988).
breakdown. Science https://doi.org/10.1126/science. healing. Wound Repair Regen. 17, 153–162 (2009). 80. Murphy, P. M. & Tiffany, H. L. Cloning of complementary
aay9792 (2020). 58. Li, Q., Park, P. W., Wilson, C. L. & Parks, W. C. DNA encoding a functional human interleukin-8
32. Pond, K. W., Doubrovinski, K. & Thorne, C. A. Matrilysin shedding of syndecan-1 regulates receptor. Science 253, 1280–1283 (1991).
Wnt/β-catenin signaling in tissue self-​organization. chemokine mobilization and transepithelial efflux of 81. Holmes, W. E., Lee, J., Kuang, W. J., Rice, G. C. &
Genes (Basel) https://doi.org/10.3390/ neutrophils in acute lung injury. Cell 111, 635–646 Wood, W. I. Structure and functional expression
genes11080939 (2020). (2002). of a human interleukin-8 receptor. Science 253,
33. Lu, P., Takai, K., Weaver, V. M. & Werb, Z. Extracellular 59. Schumann, K. et al. Immobilized chemokine fields 1278–1280 (1991).
matrix degradation and remodeling in development and soluble chemokine gradients cooperatively shape 82. Griffith, J. W., Sokol, C. L. & Luster, A. D. Chemokines
and disease. Cold Spring Harb. Perspect Biol. https:// migration patterns of dendritic cells. Immunity 32, and chemokine receptors: positioning cells for host
doi.org/10.1101/cshperspect.a005058 (2011). 703–713 (2010). defense and immunity. Annu. Rev. Immunol. 32,
34. Afonso, P. V., McCann, C. P., Kapnick, S. M. & 60. Zhu, M. et al. Spatial mapping of tissue properties 659–702 (2014).
Parent, C. A. Discoidin domain receptor 2 regulates in vivo reveals a 3D stiffness gradient in the mouse 83. Hughes, C. E. & Nibbs, R. J. B. A guide to chemokines
neutrophil chemotaxis in 3D collagen matrices. Blood limb bud. Proc. Natl Acad. Sci. USA 117, 4781–4791 and their receptors. FEBS J. 285, 2944–2971 (2018).
121, 1644–1650 (2013). (2020). 84. Zlotnik, A. & Yoshie, O. Chemokines: a new
35. Shields, J. D. et al. Autologous chemotaxis as a This is a technical tour de force demonstrating classification system and their role in immunity.
mechanism of tumor cell homing to lymphatics via the existence of stiffness gradients in mouse Immunity 12, 121–127 (2000).
interstitial flow and autocrine CCR7 signaling. Cancer embryos. 85. Sallusto, F. & Baggiolini, M. Chemokines and
Cell 11, 526–538 (2007). 61. Tran, V. D. & Kumar, S. Transduction of cell and matrix leukocyte traffic. Nat. Immunol. 9, 949–952 (2008).
36. Polacheck, W. J., Charest, J. L. & Kamm, R. D. geometric cues by the actin cytoskeleton. Curr. Opin. 86. Locati, M. & Murphy, P. M. Chemokines and
Interstitial flow influences direction of tumor cell Cell Biol. 68, 64–71 (2020). chemokine receptors: biology and clinical relevance

544 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

in inflammation and AIDS. Annu. Rev. Med. 50, 113. Sitarska, E. & Diz-​Muñoz, A. Pay attention to the local alignment of adhesions and individual
425–440 (1999). to membrane tension: mechanobiology of the protrusions. Sci. Rep. 7, 14380 (2017).
87. Proudfoot, A. E. Chemokine receptors: multifaceted cell surface. Curr. Opin. Cell Biol. 66, 11–18 This is a key article showing that cells can sense the
therapeutic targets. Nat. Rev. Immunol. 2, 106–115 (2020). topology of aligned fibres even in the absence of
(2002). 114. Baschieri, F. et al. Frustrated endocytosis controls myosin activity.
88. He, H. Q. & Ye, R. D. The formyl peptide contractility-​independent mechanotransduction at 134. Lou, H. Y. et al. Membrane curvature underlies actin
receptors: diversity of ligands and mechanism for clathrin-​coated structures. Nat. Commun. 9, 3825 reorganization in response to nanoscale surface
recognition. Molecules https://doi.org/10.3390/ (2018). topography. Proc. Natl Acad. Sci. USA 116,
molecules22030455 (2017). 115. Douguet, D. & Honoré, E. Mammalian 23143–23151 (2019).
89. Yokomizo, T. Two distinct leukotriene B4 receptors, mechanoelectrical transduction: structure and This is an important article identifying membrane
BLT1 and BLT2. J. Biochem. 157, 65–71 (2015). function of force-​gated ion channels. Cell 179, curvature-​sensing BAR-​family protein FBP17
90. Sadik, C. D., Kim, N. D. & Luster, A. D. Neutrophils 340–354 (2019). as the bridge between surface topology and
cascading their way to inflammation. Trends Immunol. 116. Goult, B. T., Yan, J. & Schwartz, M. A. Talin as a topology-​induced reorganization of the actin
32, 452–460 (2011). mechanosensitive signaling hub. J. Cell Biol. 217, network.
91. Anand-​Apte, B. & Zetter, B. Signaling mechanisms in 3776–3784 (2018). 135. McGregor, A. L., Hsia, C. R. & Lammerding, J. Squish
growth factor-​stimulated cell motility. Stem Cells 15, 117. Grashoff, C. et al. Measuring mechanical tension and squeeze-​the nucleus as a physical barrier during
259–267 (1997). across vinculin reveals regulation of focal adhesion migration in confined environments. Curr. Opin. Cell
92. Vorotnikov, A. V. & Tyurin-​Kuzmin, P. A. Chemotactic dynamics. Nature 466, 263–266 (2010). Biol. 40, 32–40 (2016).
signaling in mesenchymal cells compared to amoeboid This seminal article establishes a tension-​based 136. Lomakin, A. J. et al. The nucleus acts as a ruler
cells. Genes Dis. 1, 162–173 (2014). Förster resonance energy transfer biosensor using tailoring cell responses to spatial constraints. Science
93. Bear, J. E. & Haugh, J. M. Directed migration the focal adhesion protein vinculin. https://doi.org/10.1126/science.aba2894 (2020).
of mesenchymal cells: where signaling and the 118. Sawada, Y. et al. Force sensing by mechanical 137. Venturini, V. et al. The nucleus measures shape
cytoskeleton meet. Curr. Opin. Cell Biol. 30, 74–82 extension of the Src family kinase substrate p130Cas. changes for cellular proprioception to control dynamic
(2014). Cell 127, 1015–1026 (2006). cell behavior. Science https://doi.org/10.1126/science.
94. Gorla, M. & Bashaw, G. J. Molecular mechanisms 119. MacKay, J. L. & Hammer, D. A. Stiff substrates aba2644 (2020).
regulating axon responsiveness at the midline. enhance monocytic cell capture through E-​selectin but This article, along with Lomakin et al. (2020),
Dev. Biol. 466, 12–21 (2020). not P-​selectin. Integr. Biol. 8, 62–72 (2016). shows that cells interpret cell shape and control
95. Bellon, A. & Mann, F. Keeping up with advances in 120. Sun, X. et al. Mechanosensing through direct binding movement in confined spaces by gauging nuclear
axon guidance. Curr. Opin. Neurobiol. 53, 183–191 of tensed F-​Actin by LIM domains. Dev. Cell 55, deformation and initiating signalling events at
(2018). 468–482.e467 (2020). the nucleus.
96. Tessier-​Lavigne, M. & Goodman, C. S. The molecular 121. Winkelman, J. D., Anderson, C. A., Suarez, C., 138. Zhao, M. Electrical fields in wound healing — an
biology of axon guidance. Science 274, 1123–1133 Kovar, D. R. & Gardel, M. L. Evolutionarily diverse overriding signal that directs cell migration.
(1996). LIM domain-​containing proteins bind stressed actin Semin. Cell Dev. Biol. 20, 674–682 (2009).
97. Mirakaj, V. & Rosenberger, P. Immunomodulatory filaments through a conserved mechanism. Proc. Natl 139. Allen, G. M., Mogilner, A. & Theriot, J. A.
functions of neuronal guidance proteins. Trends Acad. Sci. USA 117, 25532–25542 (2020). Electrophoresis of cellular membrane components
Immunol. 38, 444–456 (2017). This article, along with Sun et al. (2020), creates the directional cue guiding keratocyte
98. Wu, J. Y. et al. The neuronal repellent Slit inhibits shows that LIM domain-​containing proteins galvanotaxis. Curr. Biol. 23, 560–568 (2013).
leukocyte chemotaxis induced by chemotactic factors. bind differentially to actin filaments based on This is a key article establishing the mechanism
Nature 410, 948–952 (2001). filament tension. of galvanotaxis.
99. Tole, S. et al. The axonal repellent, Slit2, inhibits 122. Rong, Y. et al. The Golgi microtubules regulate single 140. Sarkar, A., Kobylkevich, B. M., Graham, D. M.
directional migration of circulating neutrophils. cell durotaxis. EMBO Rep. https://doi.org/10.15252/ & Messerli, M. A. Electromigration of cell surface
J. Leukoc. Biol. 86, 1403–1415 (2009). embr.202051094 (2021). macromolecules in DC electric fields during cell
100. Pilling, D., Chinea, L. E., Consalvo, K. M. & Gomer, R. H. 123. Guilluy, C. et al. Isolated nuclei adapt to force and polarization and galvanotaxis. J. Theor. Biol. 478,
Different isoforms of the neuronal guidance molecule reveal a mechanotransduction pathway in the nucleus. 58–73 (2019).
Slit2 directly cause chemoattraction or chemorepulsion Nat. Cell Biol. 16, 376–381 (2014). 141. Lin, B. J. et al. Lipid rafts sense and direct electric
of human neutrophils. J. Immunol. 202, 239–248 124. Arsenovic, P. T. et al. Nesprin-2G, a component of the field-​induced migration. Proc. Natl Acad. Sci. USA
(2019). nuclear LINC complex, is subject to myosin-​dependent 114, 8568–8573 (2017).
101. Huttenlocher, A. & Horwitz, A. R. Integrins in cell tension. Biophys. J. 110, 34–43 (2016). 142. Li, X., Miao, Y., Pal, D. S. & Devreotes, P. N.
migration. Cold Spring Harb. Perspect Biol. 3, 125. Gomes, E. R., Jani, S. & Gundersen, G. G. Nuclear Excitable networks controlling cell migration during
a005074 (2011). movement regulated by Cdc42, MRCK, myosin, and development and disease. Semin. Cell. Dev. Biol. 100,
102. Henning Stumpf, B., Ambriovic´-Ristov, A., Radenovic, A. actin flow establishes MTOC polarization in migrating 133–142 (2020).
& Smith, A. S. Recent advances and prospects in the cells. Cell 121, 451–463 (2005). 143. Xiao, Z., Zhang, N., Murphy, D. B. & Devreotes, P. N.
research of nascent adhesions. Front. Physiol. 11, 126. Petrie, R. J., Koo, H. & Yamada, K. M. Generation Dynamic distribution of chemoattractant receptors
574371 (2020). of compartmentalized pressure by a nuclear piston in living cells during chemotaxis and persistent
103. Schwarz, J. et al. Dendritic cells interpret haptotactic governs cell motility in a 3D matrix. Science 345, stimulation. J. Cell Biol. 139, 365–374 (1997).
chemokine gradients in a manner governed by signal-​ 1062–1065 (2014). 144. Servant, G., Weiner, O. D., Neptune, E. R., Sedat, J. W.
to-noise ratio and dependent on GRK6. Curr Biol 27, 127. Lou, H. Y., Zhao, W., Zeng, Y. & Cui, B. The role of & Bourne, H. R. Dynamics of a chemoattractant
1314–1325 (2017). membrane curvature in nanoscale topography-​induced receptor in living neutrophils during chemotaxis.
This study shows differential regulation of the intracellular signaling. Acc. Chem. Res. 51, Mol. Biol. Cell 10, 1163–1178 (1999).
same chemokine receptor in the context of a 1046–1053 (2018). 145. Nieto, M. et al. Polarization of chemokine receptors
chemotactic and haptotactic cue. 128. Driscoll, M. K., Sun, X., Guven, C., Fourkas, J. T. & to the leading edge during lymphocyte chemotaxis.
104. Shellard, A. & Mayor, R. Durotaxis: the hard path Losert, W. Cellular contact guidance through dynamic J. Exp. Med. 186, 153–158 (1997).
from in vitro to in vivo. Dev. Cell https://doi.org/ sensing of nanotopography. ACS Nano 8, 3546–3555 146. Bailly, M. et al. Epidermal growth factor receptor
10.1016/j.devcel.2020.11.019 (2020). (2014). distribution during chemotactic responses. Mol. Biol.
105. Sunyer, R. & Trepat, X. Durotaxis. Curr. Biol. 30, 129. Chen, S. et al. Actin cytoskeleton and focal adhesions Cell 11, 3873–3883 (2000).
R383–r387 (2020). regulate the biased migration of breast cancer cells 147. Bergeron, J. J., Di Guglielmo, G. M., Dahan, S.,
106. Janmey, P. A., Fletcher, D. A. & Reinhart-​King, C. A. on nanoscale asymmetric sawteeth. ACS Nano 13, Dominguez, M. & Posner, B. I. Spatial and temporal
Stiffness sensing by cells. Physiol. Rev. 100, 695–724 1454–1468 (2019). regulation of receptor tyrosine kinase activation and
(2020). This article shows that breast cancer cell lines intracellular signal transduction. Annu. Rev. Biochem.
107. Astrof, N. S., Salas, A., Shimaoka, M., Chen, J. exhibit different migration phenotypes on 85, 573–597 (2016).
& Springer, T. A. Importance of force linkage in asymmetric sawtooth nanopatterns through the 148. Callan-​Jones, A. C. & Voituriez, R. Actin flows in
mechanochemistry of adhesion receptors. distinct regulation of actin polymerization waves cell migration: from locomotion and polarity to
Biochemistry 45, 15020–15028 (2006). and focal adhesion dynamics. trajectories. Curr. Opin. Cell Biol. 38, 12–17 (2016).
108. Jiang, G., Huang, A. H., Cai, Y., Tanase, M. & 130. Le Maout, E., Lo Vecchio, S., Kumar Korla, P., 149. Parent, C. A., Blacklock, B. J., Froehlich, W. M.,
Sheetz, M. P. Rigidity sensing at the leading edge Jinn-​Chyuan Sheu, J. & Riveline, D. Ratchetaxis Murphy, D. B. & Devreotes, P. N. G protein signaling
through alphavbeta3 integrins and RPTPalpha. in channels: entry point and local asymmetry set events are activated at the leading edge of
Biophys. J. 90, 1804–1809 (2006). cell directions in confinement. Biophys. J. 119, chemotactic cells. Cell 95, 81–91 (1998).
109. Nordenfelt, P. et al. Direction of actin flow dictates 1301–1308 (2020). 150. Servant, G. et al. Polarization of chemoattractant
integrin LFA-1 orientation during leukocyte migration. This article shows how cell direction through 3D receptor signaling during neutrophil chemotaxis.
Nat. Commun. 8, 2047 (2017). ratchet channels is dictated by spatial asymmetry Science 287, 1037–1040 (2000).
110. Moore, T. I., Aaron, J., Chew, T. L. & Springer, T. A. in two-​sided confined channels and the entry point 151. Haugh, J. M., Codazzi, F., Teruel, M. & Meyer, T.
Measuring integrin conformational change on the cell of the all-​sided confined channels. Spatial sensing in fibroblasts mediated by 3’
surface with super-​resolution microscopy. Cell Rep. 131. Ramirez-​San Juan, G. R., Oakes, P. W. & Gardel, M. L. phosphoinositides. J. Cell Biol. 151, 1269–1280
22, 1903–1912 (2018). Contact guidance requires spatial control of leading-​ (2000).
111. Lämmermann, T. et al. Rapid leukocyte migration by edge protrusion. Mol. Biol. Cell 28, 1043–1053 152. Artemenko, Y., Lampert, T. J. & Devreotes, P. N.
integrin-​independent flowing and squeezing. Nature (2017). Moving towards a paradigm: common mechanisms
453, 51–55 (2008). 132. Jang, K. J. et al. Two distinct filopodia populations at of chemotactic signaling in Dictyostelium and
112. Hetmanski, J. H. R. et al. Membrane tension the growth cone allow to sense nanotopographical mammalian leukocytes. Cell Mol. Life Sci. 71,
orchestrates rear retraction in matrix-​directed cell extracellular matrix cues to guide neurite outgrowth. 3711–3747 (2014).
migration. Dev. Cell 51, 460–475.e410 (2019). PLoS ONE 5, e15966 (2010). 153. Kunisaki, Y. et al. DOCK2 is a Rac activator that
This recent study shows the importance of 133. Kubow, K. E., Shuklis, V. D., Sales, D. J. & Horwitz, A. R. regulates motility and polarity during neutrophil
membrane tension in directed migration. Contact guidance persists under myosin inhibition due chemotaxis. J. Cell Biol. 174, 647–652 (2006).

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 545

0123456789();:
Reviews

154. Zhao, T. et al. Signaling requirements for translocation 179. Rothenberg, K. E., Scott, D. W., Christoforou, N. 200. Davidson, A. J., Amato, C., Thomason, P. A. &
of P-​Rex1, a key Rac2 exchange factor involved & Hoffman, B. D. Vinculin force-​sensitive dynamics Insall, R. H. WASP family proteins and formins
in chemoattractant-​stimulated human neutrophil at focal adhesions enable effective directed cell compete in pseudopod- and bleb-​based migration.
function. J. Leukoc. Biol. 81, 1127–1136 (2007). migration. Biophys. J. 114, 1680–1694 (2018). J. Cell Biol. 217, 701–714 (2018).
155. Yan, J. et al. A Gβγ effector, ElmoE, transduces GPCR This recent work using the vinculin Förster 201. Vargas, P. et al. Innate control of actin nucleation
signaling to the actin network during chemotaxis. resonance energy transfer-​based tension sensor determines two distinct migration behaviours in
Dev. Cell 22, 92–103 (2012). shows precise regulation of tension at different dendritic cells. Nat. Cell Biol. 18, 43–53 (2016).
156. Asokan, S. B. et al. Mesenchymal chemotaxis requires focal adhesions during directed migration. 202. Menon, S. et al. The E3 ubiquitin ligase TRIM9 is a
selective inactivation of myosin II at the leading edge 180. Ringer, P. et al. Multiplexing molecular tension sensors filopodia off switch required for netrin-​dependent axon
via a noncanonical PLCgamma/PKCalpha pathway. reveals piconewton force gradient across talin-1. guidance. Dev. Cell 35, 698–712 (2015).
Dev. Cell 31, 747–760 (2014). Nat Methods 14, 1090–1096 (2017). 203. Oakes, P. W. et al. Lamellipodium is a myosin-​
157. Hoeller, O. & Kay, R. R. Chemotaxis in the absence 181. LaCroix, A. S., Lynch, A. D., Berginski, M. E. & independent mechanosensor. Proc. Natl Acad.
of PIP3 gradients. CB 17, 813–817 (2007). Hoffman, B. D. Tunable molecular tension sensors Sci. USA 115, 2646–2651 (2018).
158. Liu, L., Das, S., Losert, W. & Parent, C. A. mTORC2 reveal extension-​based control of vinculin loading. 204. Wong, S., Guo, W. H. & Wang, Y. L. Fibroblasts probe
regulates neutrophil chemotaxis in a cAMP- and eLife https://doi.org/10.7554/eLife.33927 (2018). substrate rigidity with filopodia extensions before
RhoA-​dependent fashion. Dev. Cell 19, 845–857 182. Huang, D. L., Bax, N. A., Buckley, C. D., Weis, W. I. & occupying an area. Proc. Natl Acad. Sci. USA 111,
(2010). Dunn, A. R. Vinculin forms a directionally asymmetric 17176–17181 (2014).
159. Chen, M. Y., Long, Y. & Devreotes, P. N. A novel catch bond with F-​actin. Science 357, 703–706 205. Andrew, N. & Insall, R. H. Chemotaxis in shallow
cytosolic regulator, Pianissimo, is required for (2017). gradients is mediated independently of PtdIns
chemoattractant receptor and G protein-​mediated 183. Owen, L. M., Bax, N. A., Weis, W. I. & Dunn, A. R. 3-kinase by biased choices between random
activation of the 12 transmembrane domain adenylyl The C-​terminal actin binding domain of talin forms protrusions. Nat. Cell Biol. 9, 193–200 (2007).
cyclase in Dictyostelium. Genes Dev. 11, 3218–3231 an asymmetric catch bond with F-​actin. Preprint at 206. Welf, E. S., Ahmed, S., Johnson, H. E., Melvin, A. T.
(1997). bioRxiv https://doi.org/10.1101/2020.09.01.276568 & Haugh, J. M. Migrating fibroblasts reorient
160. Lee, S. et al. TOR complex 2 integrates cell movement (2020). directionality by a metastable, PI3K-​dependent
during chemotaxis and signal relay in Dictyostelium. 184. Singh, A. V. et al. Astrocytes increase ATP exocytosis mechanism. J. Cell Biol. 197, 105–114 (2012).
Mol. Biol. Cell 16, 4572–4583 (2005). mediated calcium signaling in response to microgroove 207. Sun, X. et al. Asymmetric nanotopography biases
161. He, Y. et al. Mammalian target of rapamycin and structures. Sci. Rep. 5, 7847 (2015). cytoskeletal dynamics and promotes unidirectional
Rictor control neutrophil chemotaxis by regulating 185. Hung, W. C. et al. Confinement sensing and signal cell guidance. Proc. Natl Acad. Sci. USA 112,
Rac/Cdc42 activity and the actin cytoskeleton. optimization via Piezo1/PKA and myosin II pathways. 12557–12562 (2015).
Mol. Biol. Cell 24, 3369–3380 (2013). Cell Rep. 15, 1430–1441 (2016). 208. Vicente-​Manzanares, M., Ma, X., Adelstein, R. S. &
162. Chen, L. et al. PLA2 and PI3K/PTEN pathways act 186. Bavi, N., Richardson, J., Heu, C., Martinac, B. & Horwitz, A. R. Non-​muscle myosin II takes centre stage
in parallel to mediate chemotaxis. Dev. Cell 12, Poole, K. PIEZO1-mediated currents are modulated in cell adhesion and migration. Nat. Rev. Mol. Cell
603–614 (2007). by substrate mechanics. ACS Nano 13, 13545–13559 Biol. 10, 778–790 (2009).
163. van Haastert, P. J., Keizer-​Gunnink, I. & Kortholt, A. (2019). 209. Plotnikov, S. V., Pasapera, A. M., Sabass, B. &
Essential role of PI3-kinase and phospholipase A2 This article shows that the surface geometry, Waterman, C. M. Force fluctuations within focal
in Dictyostelium discoideum chemotaxis. J. Cell Biol. including roughness and stiffness, modulates adhesions mediate ECM-​rigidity sensing to guide
177, 809–816 (2007). PIEZO1 channel activation in response to the directed cell migration. Cell 151, 1513–1527
164. Heit, B. et al. PTEN functions to ‘prioritize’ chemotactic external mechanical force applied at cell–substrate (2012).
cues and prevent ‘distraction’ in migrating neutrophils. interfaces. 210. Chan, K. T., Bennin, D. A. & Huttenlocher, A.
Nat. Immunol. 9, 743–752 (2008). 187. Subramanian, B. C. et al. The LTB4-BLT1 axis Regulation of adhesion dynamics by calpain-​mediated
165. Meliton, A. Y. et al. Cytosolic group IVa phospholipase regulates actomyosin and β2-integrin dynamics during proteolysis of focal adhesion kinase (FAK). J. Biol.
A2 mediates IL-8/CXCL8-induced transmigration neutrophil extravasation. J. Cell Biol. https://doi.org/ Chem. 285, 11418–11426 (2010).
of human polymorphonuclear leukocytes in vitro. 10.1083/jcb.201910215 (2020). 211. Mayor, R. & Etienne-​Manneville, S. The front and rear
J. Inflamm. 7, 14 (2010). This article reports a critical role for neutrophil-​ of collective cell migration. Nat. Rev. Mol. Cell Biol.
166. Yonker, L. M. et al. Neutrophil-​derived cytosolic derived LTB4 in the regulation of non-​muscle 17, 97–109 (2016).
PLA2α contributes to bacterial-​induced neutrophil myosin II and β2 integrin that depends on the 212. Ladoux, B. & Mège, R. M. Mechanobiology of
transepithelial migration. J. Immunol. 199, release of extracellular vesicles during neutrophil collective cell behaviours. Nat. Rev. Mol. Cell Biol. 18,
2873–2884 (2017). arrest and extravasation in live mice. 743–757 (2017).
167. Ma, H., Gamper, M., Parent, C. & Firtel, R. A. The 188. Gao, R. et al. A large-​scale screen reveals genes that 213. Shellard, A., Szabó, A., Trepat, X. & Mayor, R.
Dictyostelium MAP kinase kinase DdMEK1 regulates mediate electrotaxis in Dictyostelium discoideum. Supracellular contraction at the rear of neural crest
chemotaxis and is essential for chemoattractant-​ Sci. Signal 8, ra50 (2015). cell groups drives collective chemotaxis. Science 362,
mediated activation of guanylyl cyclase. EMBO J. 16, 189. Sánchez, M. F., Els-​Heindl, S., Beck-​Sickinger, A. G., 339–343 (2018).
4317–4332 (1997). Wieneke, R. & Tampé, R. Photo-​induced receptor This article shows a rear wheel drive-​like motility
168. Mendoza, M. C., Booth, E. O., Shaulsky, G. & confinement drives ligand-​independent GPCR during collective chemotaxis by actomyosin
Firtel, R. A. MEK1 and protein phosphatase 4 signaling. Science https://doi.org/10.1126/science. contractility in a cluster of rear cells.
coordinate Dictyostelium development and abb7657 (2021). 214. Yam, P. T. et al. Actin-​myosin network reorganization
chemotaxis. Mol. Cell Biol. 27, 3817–3827 (2007). 190. Smith, H. E. Nematode sperm motility. WormBook breaks symmetry at the cell rear to spontaneously
169. Heit, B., Tavener, S., Raharjo, E. & Kubes, P. https://doi.org/10.1895/wormbook.1.68.2 (2014). initiate polarized cell motility. J. Cell Biol. 178,
An intracellular signaling hierarchy determines 191. Pollard, T. D. & Borisy, G. G. Cellular motility driven by 1207–1221 (2007).
direction of migration in opposing chemotactic assembly and disassembly of actin filaments. Cell 112, 215. Tsai, T. Y. et al. Efficient front-​rear coupling in
gradients. J. Cell Biol. 159, 91–102 (2002). 453–465 (2003). neutrophil chemotaxis by dynamic myosin II
170. Liu, X. et al. Bidirectional regulation of neutrophil 192. Bodor, D. L., Pönisch, W., Endres, R. G. & Paluch, E. K. localization. Dev. Cell 49, 189–205.e186 (2019).
migration by mitogen-​activated protein kinases. Of cell shapes and motion: the physical basis of animal This article shows that the side-​to-side
Nat. Immunol. 13, 457–464 (2012). cell migration. Dev. Cell 52, 550–562 (2020). redistribution of myosin II at the back of cells in
171. Mouneimne, G. et al. Spatial and temporal control of 193. Krause, M. & Gautreau, A. Steering cell migration: response to leading-​edge turning is critical for
cofilin activity is required for directional sensing during lamellipodium dynamics and the regulation of maintaining persistence and turning ability in
chemotaxis. Curr. Biol. 16, 2193–2205 (2006). directional persistence. Nat. Rev. Mol. Cell Biol. 15, neutrophils undergoing chemotaxis.
172. Miao, Y. et al. Altering the threshold of an excitable 577–590 (2014). 216. Allen, G. M. et al. Cell mechanics at the rear act to
signal transduction network changes cell migratory 194. McCormick, L. E. & Gupton, S. L. Mechanistic steer the direction of cell migration. Cell Syst. 11,
modes. Nat. Cell Biol. 19, 329–340 (2017). advances in axon pathfinding. Curr. Opin. Cell Biol. 286–299.e284 (2020).
173. Zhan, H. et al. An excitable Ras/PI3K/ERK 63, 11–19 (2020). 217. Charras, G. & Paluch, E. Blebs lead the way: how to
signaling network controls migration and oncogenic 195. Johnson, H. E. et al. F-​actin bundles direct the migrate without lamellipodia. Nat. Rev. Mol. Cell Biol.
transformation in epithelial cells. Dev. Cell 54, initiation and orientation of lamellipodia through 9, 730–736 (2008).
608–623.e605 (2020). adhesion-​based signaling. J. Cell Biol. 208, 443–455 218. Zatulovskiy, E., Tyson, R., Bretschneider, T. & Kay, R. R.
174. King, S. J. et al. Lamellipodia are crucial for (2015). Bleb-​driven chemotaxis of Dictyostelium cells. J. Cell
haptotactic sensing and response. J. Cell Sci. 129, 196. Svitkina, T. M. et al. Mechanism of filopodia initiation Biol. 204, 1027–1044 (2014).
2329–2342 (2016). by reorganization of a dendritic network. J. Cell Biol. 219. Reversat, A. et al. Cellular locomotion using
175. Cox, C. D., Bavi, N. & Martinac, B. Biophysical 160, 409–421 (2003). environmental topography. Nature 582, 582–585
principles of ion-​channel-mediated mechanosensory 197. Wu, C. et al. Arp2/3 is critical for lamellipodia (2020).
transduction. Cell Rep. 29, 1–12 (2019). and response to extracellular matrix cues but is This article identifies friction force generated by
176. Pal, N., Wu, M. & Lu, H. P. Probing conformational dispensable for chemotaxis. Cell 148, 973–987 retrograde flow of actin propelling cells forward
dynamics of an enzymatic active site by an in situ (2012). through a 3D environment in the absence of
single fluorogenic probe under piconewton force This is the first article to establish that the Arp2/3 integrin engagement.
manipulation. Proc. Natl Acad. Sci. USA 113, complex is required for ECM haptotaxis but is 220. Aoun, L. et al. Amoeboid swimming is propelled by
15006–15011 (2016). dispensable for RTK-​dependent chemotaxis in molecular paddling in lymphocytes. Biophys. J. 119,
177. Hu, X., Margadant, F. M., Yao, M. & Sheetz, M. P. fibroblasts. 1157–1177 (2020).
Molecular stretching modulates mechanosensing 198. Schaks, M., Giannone, G. & Rottner, K. Actin dynamics This fascinating recent work demonstrates a new
pathways. Protein Sci. 26, 1337–1351 (2017). in cell migration. Essays Biochem. 63, 483–495 mechanism of cell swimming using ‘molecular
178. Kumar, A. et al. Talin tension sensor reveals novel (2019). paddling’ of cell surface proteins coupled to flow
features of focal adhesion force transmission and 199. Collins, S. R. et al. Using light to shape chemical of the underlying cell cortex.
mechanosensitivity. J. Cell Biol. 213, 371–383 gradients for parallel and automated analysis of 221. Purcell, E. M. Life at low Reynolds number. Am. J.
(2016). chemotaxis. Mol. Syst. Biol. 11, 804 (2015). Phys. 45, 3–11 (1977).

546 | August 2021 | volume 22 www.nature.com/nrm

0123456789();:
Reviews

222. Yamada, K. M. & Sixt, M. Mechanisms of 3D cell 229. Haeger, A., Wolf, K., Zegers, M. M. & Friedl, P. Acknowledgements
migration. Nat. Rev. Mol. Cell Biol. 20, 738–752 Collective cell migration: guidance principles The authors apologize to their colleagues whose work on this
(2019). and hierarchies. Trends Cell Biol. 25, 556–566 vast topic they were unable to cite due to length constraints.
223. Dai, W. et al. Tissue topography steers migrating (2015). They thank M. Butler for designing some of the artwork in
Drosophila border cells. Science 370, 987–990 230. Lämmermann, T. & Sixt, M. Mechanical modes of Fig. 3c, and J. Haugh for thoughtful discussions. J.E.B.
(2020). ‘amoeboid’ cell migration. Curr. Opin. Cell Biol. 21, acknowledges support from the NIH (R35GM130312 and
This article shows that while chemical cues promote 636–644 (2009). U01EB018816). C.A.P. acknowledges support from the NIH
posterior movement, topographical cues provide 231. Pagès, D.-L. et al. Cell clusters adopt a (R01AI152517).
orthogonal information and a path of least collective amoeboid mode of migration in
resistance during the migration of border cells confined non-​adhesive environments. Preprint at Author contributions
in the egg chamber of D. melanogaster embryos. bioRxiv https://doi.org/10.1101/2020.05.28.106203 The authors contributed equally to all aspects of the article.
224. Mathias, J. R. et al. Resolution of inflammation by (2020).
retrograde chemotaxis of neutrophils in transgenic 232. Liu, Y. J. et al. Confinement and low adhesion induce Competing interests
zebrafish. J. Leukoc. Biol. 80, 1281–1288 (2006). fast amoeboid migration of slow mesenchymal cells. The authors declare no competing interests.
225. Wang, J. et al. Visualizing the function and fate of Cell 160, 659–672 (2015).
neutrophils in sterile injury and repair. Science 358, 233. Ruprecht, V. et al. Cortical contractility triggers a Peer review information
111–116 (2017). stochastic switch to fast amoeboid cell motility. Nature Reviews Molecular Cell Biology thanks R. Insall, M.
226. Hind, L. E. & Huttenlocher, A. Neutrophil reverse Cell 160, 673–685 (2015). Sixt and the other, anonymous, reviewer(s) for their contribution
migration and a chemokinetic resolution. Dev. Cell 47, 234. Graziano, B. R. et al. Cell confinement reveals a to the peer review of this work.
404–405 (2018). branched-​actin independent circuit for neutrophil
227. Paluch, E. K., Aspalter, I. M. & Sixt, M. Focal polarity. PLoS Biol. 17, e3000457 (2019). Publisher’s note
adhesion-​independent cell migration. Annu. Rev. Cell 235. Lehmann, S. et al. Hypoxia induces a HIF-1-dependent Springer Nature remains neutral with regard to jurisdictional
Dev. Biol. 32, 469–490 (2016). transition from collective-​to-amoeboid dissemination claims in published maps and institutional affiliations.
228. Shellard, A. & Mayor, R. All roads lead to directional in epithelial cancer cells. Curr. Biol. 27, 392–400
cell migration. Trends Cell Biol. 30, 852–868 (2020). (2017). © Springer Nature Limited 2021

NAture RevIews | MolECular CEll BioloGy volume 22 | August 2021 | 547

0123456789();:

You might also like